PDF hosted at the Radboud Repository of the Radboud University Nijmegen The following full text is a publisher's version. For additional information about this publication click this link. http://hdl.handle.net/2066/145089 Please be advised that this information was generated on 2017-06-17 and may be subject to change. Chronobiology of absence epilepsy Magdalena Kinga Smyk © Magdalena Kinga Smyk ISBN: 978-94-6284-031-7 Printed in Kraków Chronobiology of absence epilepsy Proefschrift ter verkrijging van de graad van doctor aan de Radboud Universiteit Nijmegen op gezag van de rector magnificus prof. dr. Th.L.M. Engelen, volgens besluit van het college van decanen in het openbaar te verdedigen op maandag 26 oktober 2015 om 14.30 uur precies door Magdalena Kinga Smyk geboren op 7 oktober 1983 te Rzeszów (Polen) Promotoren: Prof. dr. E.L.J.M. van Luijtelaar Prof. dr. M.H. Lewandowski (Uniwersytet Jagielloński w Krakowie, Polen) Manuscriptcommissie: Prof. dr. G. Flik Prof. dr. J.H. Meijer (Universiteit Leiden) Prof. dr. G. Hess (Uniwersytet Jagielloński w Krakowie, Polen) Chronobiology of absence epilepsy Doctoral Thesis to obtain the degree of doctor from Radboud University Nijmegen on the authority of the Rector Magnificus prof. dr. Th.L.M. Engelen, according to the decision of the Council of Deans to be defended in public on Monday, October 26, 2015 at 14.30 hours by Magdalena Kinga Smyk Born on October 7, 1983 in Rzeszów (Poland) Supervisors: Prof. dr. E.L.J.M. van Luijtelaar Prof. dr. M.H. Lewandowski (Uniwersytet Jagielloński w Krakowie , Poland) Doctoral Thesis Committee: Prof. dr. G. Flik Prof. dr. J.H. Meijer (Leiden University) Prof. dr. G. Hess (Uniwersytet Jagielloński w Krakowie, Poland) CONTENTS CHAPTER 1 General Introduction CHAPTER 2 Differential temporal organization of sleep-wake states and absence seizures in light versus dark phase in a genetic animal model 57 CHAPTER 3 Endogenous rhythm of absence epilepsy: relationship with general motor activity and sleep-wake states 87 CHAPTER 4 Internal desynchronization facilitates seizures 113 CHAPTER 5 Re-entrainment of sleep-wake states and absence seizures after a phase delay 137 CHAPTER 6 General Discussion 181 ADDENDUM English Summary Nederlandse Samenvatting Curriculum Vitae Schools and Courses Awards List of Publications Acknowledgements Podziękowania Donders Series 9 219 224 230 230 231 231 235 239 245 CHAPTER 1 General Introduction Biological timing: from milliseconds to annual cycles An accurate processing of time information requires an efficient clock. Living organisms have developed a multitude of timing mechanisms based on oscillations, unidirectional processes or a combination of these two, to perceive succession and duration of events. Perception of succession applies to the recognition of a sequential characteristic of events, their temporal order. Perception of duration means that the persistence of an event over time and the interval between two events can be identified (Fraisse, 1984; Wittman, 1999; Mattel and Meck, 2000; Rensing et al., 2001). The time scales over which organisms process temporal information and generate timed behavior encompass several orders of magnitude (Hinton and Meck, 1997; Buonomano and Karmarkar, 2002; Buonomano, 2007, Agostino et al., 2011). A microsecond scale is used by sound localization mechanisms based on the ability to discriminate between very short time intervals needed for sound to travel from one ear to the other (Buonomano, 2007). However, conscious detection of temporal order is possible when interval separating events exceed 20 – 40 milliseconds (Steinbuchel, 1995; Pastore and Farrington, 1996). This time scale, in a range of milliseconds to seconds and minutes (“interval timing”), is critical for sensory processing, motor coordination, speech recognition or associative learning (Mattel and Meck, 2000; Buonomano, 2007). Opposite to these rather fast processes, organisms use also longer intervals such as hours or days. Many biological functions occur regularly and repeat each day at precisely predictable time. Core body temperature of humans living a conventional lifestyle reaches its minimum around 5 a.m. (Waterhouse et al., 2005). Blood pressure in healthy subjects is reduced about 10 – 20% in the night in comparison to its mean daytime value (Synder et al., 1964). Human cortisol and 11 rodent corticosterone, hormones implicated in response to stress, but also in metabolic, anti-inflammatory and immunosuppressive processes and responses, reach their maximum value just before the onset of the period of daily activity: in the early morning in humans and just before the dark phase in rats (Atkinson and Waddell, 1997; Perreau-Lenz et al., 2004; Chan and Debono, 2010). Cortisol is not an exception, a variety of hormones such as melatonin, thyroidstimulating hormone (TSH), and prolactin, cycle with a period of 24 h (Czeisler et al., 2005; Schechter and Boivin, 2010). These are a few examples of many circadian rhythms (a term originated from the Latin words circa, meaning “about”, and dies meaning “day”), oscillations of biological processes representing the internal notion of time. The frequency of oscillations is one of the criteria on which rhythmic processes are classified. While the duration of circadian rhythms does not deviate significantly from 24 h, the range of ultradian (shorter than a day) and infradian (longer than a day, e.g. lunar, tidal, annual) periodicities may vary enormously. To illustrate, the period of ultradian rhythms may vary from milliseconds to hours, and that from infradian rhythms from days to years. For example, the period length of gamma oscillations as occurring in neuronal network is 0.01 – 0.03 sec, while the rapid-eye-movement (REM)/slow-wave sleep cycle in humans, both within the ultradian range, is 1.5 h (Achermann and Borbély, 1990; Achermann et al., 1993; Fries et al., 2007). Similarly, the infradian human menstrual cycle lasts about 28 days, while the reproduction cycle of seasonal breeding animals lasts about 12 months (Chiazze et al., 1968; Lee and Zucker, 1991; Woodfill et al., 1994). Besides the frequency, rhythmic phenomena may also be classified according to the biological system in which they are observed: a single cell, complex organism or a population, to their origin: endogenous or exogenous, and to their function (Aschoff, 1981). 12 Different time mechanisms in the mammalian brain are interrelated. The circadian pacemaker has been reported to affect the perception of time interval (Poppel and Giedke, 1970). Next, the seasonal-encoding mechanisms use the circadian timing system to estimate the day length; this is necessary for recognition of different seasons of the year (Goldman, 2001; Prendergast et al., 2002). Circadian timing system: from single-cell oscillators to multi-oscillatory machinery The presence of an internal mechanism providing synchronization of physiological and behavioral processes to recurrent 24 h environmental cycles was obviously advantageous from an evolutionary point of view. A circadian timing system, present already in phylogenetically early, primitive organisms such as cyanobacteria (Synechococcus elongatus), allows the anticipation and adaptation to light-dark, temperature, or predators cycles, increasing therefore chances of survival in a particular ecological niche (Pittendrigh, 1993; Ouyang et al., 1998; Woelfle et al., 2004; Hut and Beersma, 2011). The mechanism of circadian oscillations is based on complex molecular feedback loops, both positive and negative, regulating transcription of rather conservative core clock genes. Cyclic changes in clock genes expression and proteins level generate rhythmic physiological output (Reppert and Weaver, 2002; Dvornyk et al., 2003; Roenneberg and Merrow, 2003; Ko and Takahashi, 2006). The circadian clock, a self-sustained mechanism oscillating with a period length close to 24 h, needs to be entrained to external environmental time to fulfill its role. Entrainment occurs when a stable phase-relationship between the internal and external oscillations is established, that is when their period lengths are equal (Pittendrigh, 1981). By an input pathway entraining factors, called “Zeitgebers” (a 13 term originated from the German words Zeit meaning “time” and geben meaning “to give”) reach a central oscillator in which a rhythmic signal is generated and then transmitted via an output pathway to dependent oscillators, and manifests as an overt rhythm (Golombek and Rosenstein, 2010). Light is one of the strongest synchronizers for biological rhythms among the different Zeitgebers (Pittendrigh, 1960). In mammals, photic information perceived by the retina is transmitted via the retino-hypothalamic tract (RHT, input pathway) to the central circadian pacemaker: the suprachiasmatic nuclei of the hypothalamus (the SCN) (Moore and Lenn, 1972; Zordan et al., 2001). The SCN generates a rhythmic signal which, by multiple synaptic connections and humoral factors, reaches different brain areas (output pathways). As a result, rhythmic fluctuations in e.g. core body temperature, sleepwake cycle or hormone secretion are observed (Buijs and Kalsbeek, 2001). The presence of Zeitgebers is not necessary for rhythms generation considering the self-sustained character of circadian oscillations. Indeed, circadian rhythms persist in constant environmental conditions, however, in such a “free-running” state their period length deviate slightly from 24 h (Bünning, 1964; Herzog and Schwartz, 2002). Other circadian parameters such as amplitude or acrophase (the time point at which a rhythm peaks) may be also affected. The mammalian circadian timing system has a complex, hierarchical organization. Multiplicity of entraining factors, classified generally as photic and non-photic, requires a variety of input pathways (Hannibal and Fahrenkrug, 2006; Morin, 2013). Besides the photic input of the SCN from the RHT, the SCN also receives indirect input from the intergeniculate leaflet (IGL) of the lateral geniculate nucleus of the thalamus and two pretectal nuclei (posterior and olivary) (Mikkelsen and Vrang, 1994; Moore and Card, 1994; Meijer and Schwartz, 2003). All three structures are innervated by the retina. 14 Information about non-photic Zeitgebers comes from several brain structures and regions involved in arousal, sleep-wake regulation, feeding, stress responses, reproduction, olfactory, immune and autonomic functions (Hastings et al., 1997; Krout et al., 2002). The central pacemaker itself has also a complex structure. The SCN consists of about 10 000 – 15 000 densely-packed neurons oscillating with different frequencies, which all together yields a 24 h rhythm (Gillette, 1991; Welsh et al., 1995; Honma et al., 1998). The distribution of SCN cell’s phenotype with regard to neurotransmitters and direct retinal connections is not uniform in humans and in rodents (Hofmann et al., 1996; Dai et al., 1997; Morin and Allen, 2006). Besides this central master circadian pacemaker, there are numerous peripheral oscillators, both in the brain (e.g. pineal and pituitary gland, paraventricular nucleus of the hypothalamus) and in peripheral organs such as liver, lungs or skeletal muscles (Yamazaki et al., 2000; Abe et al., 2002). These peripheral clocks are in a stable phase-relationship with the SCN and the external environment. However, when the entrainment is desynchronization lost or between temporally master and disturbed, peripheral the internal (dependent) oscillators occurs. This phenomenon has been demonstrated by studies investigating the effects of phase-shifts in a light-dark cycle resembling jet lag or shift work. The amount of time required for full entrainment to a new photoperiod is not only species dependent, it also varies between different oscillators, however the master pacemaker always re-synchronizes as first (Yamazaki et al., 2000; Nagano et al., 2003; Lee et al., 2009). Lack of internal synchrony is thought to have diverse health consequences starting from relatively mild discomfort associated with feelings as often reported in jet lag, through serious sleep disturbances to depression or a higher risk of cancer (Arendt and Marks, 1982; 15 Schernhammer et al., 2001; Knutsson, 2003; Schernhammer et al., 2003; Megdal et al., 2005; Blask, 2009). Melatonin – a hand of the clock The simplified model of the circadian timing system described above assumes a linear relationship between its elements: Zeitgebers, input pathways, central oscillator and output pathways. However, there is evidence suggesting much more complex relationship between the components. Clock-dependent rhythms may give feedback to the master pacemaker or affect the input pathways (LaVail, 1980; Pfeffer et al., 2012). Also Zeitgebers themselves are able to directly influence physiological functions. This phenomenon is called “masking”. As a result of masking, the usual shape and/or parameters of a rhythm change as a consequence of random or non-random environmental stimuli. These alterations persist only for the duration of the stimulus while the endogenous components of the rhythm remain unchanged. An example of masking is the elevation of body temperature after a hot bath, direct effect of light on states of vigilance, or light-induced suppression of melatonin synthesis (Borbély et al., 1975; Bronstein et al., 1987; Aschoff, 1999). The pineal hormone melatonin is an example of a variable which, produced and secreted under the control of the central pacemaker, is able to affect the action of the central pacemaker itself. In mammals, melatonin is synthesized and released from the pineal gland into the circulation predominantly during darkness, it encodes therefore the length and the phase of the dark period, regardless of the ecological type of the animal: diurnal or nocturnal (Vanecek, 1998). Melatonin binds with high affinity to G-protein-coupled MT1 and MT2 melatonin receptors, which are distributed widely in the brain (Weaver et al., 1989). There is a high inter-species and inter-structure 16 variability in the distribution of melatonin receptors; however, two major brain regions with high presence and expression of these receptors were identified: pituitary pars tuberalis and the SCN (Weaver et al., 1989; Stankov et al., 1991; Reppert et al., 1994). The rhythmic synthesis and secretion of melatonin is one of the direct outputs of the central pacemaker, a “hand of the clock”. The melatonin rhythm is diminished after an SCN lesion or at exposure to constant bright light, as many other circadian rhythms (Perlov et al., 1981; Honma et al., 1996; Perreau-Lenz et al., 2003; Tapia-Osorio et al., 2013). On the other hand, the presence of functional melatonin receptors in the SCN makes the master pacemaker susceptible to its action. The hormone has been found to decrease the firing rate of SCN neurons, as well as to modify neuronal activity of other brain nuclei (Mason and Brooks, 1988; Naranjo-Rodriguez et al., 1991; Liu et al., 1997). This ability implicates that exogenous melatonin, given at a particular time of a day, entrains and shifts the clock (Sharkey and Eastman, 2002). The free-running activity rhythm in rodents, induced by exposure to constant environmental conditions, has been synchronized to the timing of the administration of the hormone. The entrainment was achieved mostly by advancing the onset of activity towards the injection time (Redman et al., 1983; Slotten et al., 1999; Pitrosky et al. 1999). Moreover, melatonin was reported to accelerate the speed of re-entrainment after the phase shift in the light-dark cycle (Illnerova et al., 1989; Golombek and Cardinali, 1993; Sharma et al., 1999; Hirai et al., 2005). Because of these chronobiological properties exogenous melatonin and its analogues are considered as a potential therapeutic treatment for jet lag and diseases in which circadian disturbances are present (Srinivasan et al., 2008; Spadoni et al., 2010; Srinivasan et al., 2010). 17 Rhythms description Any rhythmic process can be described by four parameters: period, mean level (MESOR), amplitude and acrophase. The period of a rhythm is the duration of a full cycle, or in other words, the distance from one peak to another. In circadian nomenclature different notations for the endogenous period of an organism and the period of the Zeitgeber are used: τ and T, respectively. There are several methods for the determination of the period starting from a simple visual inspection of the actogram (the graphical display of data along two time axes) to numerical procedures such as autocorrelation, Fourier transform, Enright (chi square) periodogram, and cosinor fit (Enright, 1965; Halberg et al., 1967; Refinetti et al., 2007). The mean of the rhythm represents a central value around which the oscillation occurs. Although arithmetic mean is commonly used for an estimation of a central tendency in statistic, rhythmadjusted mean: MESOR, yielded by the cosinor method, is more accurate and precise in the analysis of circadian rhythms especially when the data points are spaced unequally (Refinetti et al., 2007). Amplitude is defined as a one-half of the distance between the minimal and maximal value of the periodic function (Morris, 1992). If the estimation of the parameter is based on cosine fitting procedure, amplitude represents the distance between MESOR and peak or trough of the wave (Fernandez et al., 2009). Acrophase of the rhythm, a lag of the crest time of the cosine curve fitted to the data from a defined reference time point, represents the time at which a maximum value of the rhythms occurs. The acrophase can be expressed in radians, degrees or hours (Refinetti et al., 2007; Fernandez et al., 2009). Three of the parameters: MESOR, amplitude and acrophase can be computed by means of the cosinor method. The method assumes 18 that cosine curves of a known period can be fitted to the data by means of a least squares method as an estimate of the pattern of the smoothed rhythm (Halberg et al., 1967). The model can be described by the equation: xi = M + A cos (Θi + φ) + ei xi – the value of the function at time t, M – mesor, A – amplitude, Θ – angular frequency (the coefficient to translate from hours to degrees), φ – acrophase, ei – an error term assumed to be independent and normally distributed with mean zero and fixed unknown variance Figure 1. Schematic representation of cosinor analysis and circadian parameters 19 To compute mesor, amplitude and acrophase, a system of three equations can be derived and solved in algebraic form. A large advantage of the Cosinor method is that it allows the estimation of all parameters (mesor, amplitude and acrophase) at once together with confidence intervals. The method is thought to be sensitive in detection of rhythmicity even in noisy data (low signal-tonoise ratio), and superior over others such as Fourier, Enright or LombScargle periodograms (Bingham et al., 1993). There are two additional parameters used in the description of circadian rhythms: waveform, informing about the shape of the rhythm, and prominence (robustness) informing about rhythm’s strength and endurance. Prominence is represented by the percentage of the variance accounted for by the fitted cosine model, which corresponds to the coefficient of determination R 2 in regression analysis (Halberg et al., 1973). The waveform is determined according to the Cosinor method by the harmonic content (the amplitude and acrophase pairs of all harmonic terms included in the model) to account for the non-sinusoidality of the signal. Circadian rhythms in pathology and disease Circadian control upon physiological processes makes their occurrence predictable and indeed symptoms of some medical conditions tend to manifest or worsen during a particular time of a day (Smolensky and Peppas, 2007). For example, symptoms of immunerelated conditions such as allergic rhinitis or rheumatoid arthritis aggravate usually during the night or upon awaking, both periods of time coinciding with high concentration of pro-inflammatory agents (Kowanko et al., 1982; Reinberg et al., 1988; Smolensky et al., 1995; Petrovsky et al., 1998). Patients suffering from migraine attacks or depression may expect worsening of the symptoms in the early 20 morning (Wehr, 1982; Solomon, 1992). During the night, the symptoms of biliary colic and attacks of peptic ulcer are more frequent (Moore and Halberg, 1987; Rigas et al., 1990) and cardiovascular emergencies, e.g. sudden cardiac death, acute myocardial infarction or stroke, occur predominantly in the morning (Cohen et al., 1997; Elliot, 1998; Portaluppi et al., 2012). Epilepsy: circadian rhythms in the occurrence of seizures Interestingly, opposite to pathologies occurring in the cardiovascular system, symptoms of some diseases such as epilepsy show a high variability in the timing of their occurrence. A possible explanation might be that epilepsy, a neurological disorder characterized by recurrent seizures, is not a homogenous disease. It has many forms and underlying causes, which in fact may interfere differently with the circadian timing system. According to the World Health Organization (WHO) 50 million people worldwide suffer from epilepsy, 80% of the cases are found in developing regions. The incidence of the disease shows a bimodal distribution with dominant peaks in childhood and in the elderly and a slight gender-preference towards males (Kotsopoulos et al., 2002). Considering their etiology, epilepsies may be classified as structural/metabolic, genetic and of unknown origin. In the first class, termed previously “symptomatic”, the underlying cause of seizure development is associated with structural or metabolic condition such as head trauma, cerebral tumor or infection. Genetic epilepsies, previously “idiopathic”, have a well-defined or presumed genetic background with seizures being the core symptoms of the disorder (Shorvon, 2010; Berg et al., 2010). 21 Epileptic seizures, clinical manifestation of which may vary from general convulsions to brief moments of reduced consciousness without any motor signs, result from abnormal excessive or synchronous neuronal activity in the brain (Fisher, 2005). In general, seizures may be classified as focal, they have their origin in a network restricted to one hemisphere (usually to a small area in a particular lobe), and generalized, then they have their origin at some point within, rapidly engaging bilaterally distributed networks (Berg et al., 2010). The recent, simplified classification of seizures recommended by the International League Against Epilepsy (ILEA) is presented in Table 1. Classification of seizures Generalized seizures Tonic-clonic (in any combination) Absence Typical Atypical Absence with special features Myoclonic absence Eyelid myoclonia Myoclonic Myoclonic Myoclonic atonic Myoclonic tonic Clonic Tonic Atonic Focal seizures Unknown Epileptic spasm Table 1. ILAE classification of seizures. Berg et al., 2010 22 A vast variety of seizure types, their location and underlying mechanisms may contribute to a different circadian pattern of their occurrence. A non-uniform distribution of convulsive seizures was first described by Gowers (1885), who reported 21% of patients having seizures exclusively during the night, 43% during the day, and 37% of patients, whose seizures showed no preference for a particular time of the day, appearing both during the day as well as during the night. These results were then confirmed by Langdon-Down and Brain (1929) who classified seizures as diurnal, nocturnal and diffuse. More recent studies have revealed that seizures follow a distinct temporal pattern depending on the location of the focus. Epileptic activity starting in the temporal lobe (temporal lobe epilepsy, TLE) is most pronounced during the day, while epileptic activity starting in the frontal (frontal lobe epilepsy, FLE) and parietal lobe occurs more frequently during the night (Quigg et al., 1998; Quigg and Straume, 2000; Pavlova et al., 2004; Durazzo et al., 2008; Hofstra et al., 2009; Karafin et al., 2010; Zarowski et al., 2010; Loddenkemper et al., 2011). Another important factor contributing to a non-uniform occurrence of seizures across the 24 h day is the state of vigilance. TLE seizures tend to occur during wakefulness, while FLE seizures during sleep. Focal seizures may become generalized seizures and this happens more often during sleep. Also during sleep (stages I and II of slow-wave sleep), the majority of generalized seizures with spike-wave pattern are recorded (Kellaway et al., 1980; Halasz, 1991; Bazil and Walczak, 1997; Crespel et al., 2000; Pavlova et al., 2004; Sinha et al., 2006; Hofstra et al., 2009; Ramgopal, 2012). Human studies analyzing the diurnal variation of seizures and the relationship between seizures and states of vigilance are presented in Table 2. The majority of the studies describing the daily rhythms in epileptic activity was conducted in medical facilities, with strictly controlled environmental conditions providing entrainment. However, 23 to confirm a true circadian nature of a rhythmic process, it is necessary to exclude the presence of Zeitgebers. Two studies to date addressed the issue, and only one used humans with epilepsy. Pavlova and coworkers investigated the putative circadian variation of interictal epileptiform discharges (IED) in patients subjected to a forced desynchrony protocol, a method in which all behaviors are evenly scheduled across all circadian phases with environmental conditions maintained constant. In this pilot study circadian variation in IED independently of sleep-wake states was found, although in a small number of patients with a high variability in the phase of IED no circadian rhythm could be established (Pavlova et al., 2009). The group of Quigg investigated the occurrence of seizures in a rat model of TLE. Animals were kept in 12:12 light-dark cycle and then transferred to constant darkness. In this condition, a circadian rhythm in the occurrence of seizures was preserved and this proved its endogenous origin (Quigg et al., 2000). 24 Authors Design Kellaway et al. R (1980) N (seizures) Sleep/wake 19 adult, SWDs more children frequent in (N/A) SWS than in Diurnal pattern EEG N/A S S wake and REM Martins da Silva R et al. (1984) 17 adults IEDs more IEDs peak 23:00 – (N/A) frequent 07:00 and 09:30 – during SWS 11:00 than in REM Horita et al. R (1991) 4 children SWDs most SWDs concentrated in (N/A) frequent the last third of during SWS nocturnal sleep S stage I Bazil and R Walczak (1997) 188 adults 37% FLE (1116) sleep, 74% N/A S N/A S LTLE, XTLE occur S TLE wake Crespel et al. R (1998) 30 adults 61% FLE (133) sleep, 89% TLE wake Quigg et al. R (1998) 98 adults N/A (1350) randomly, MTLE peak at 15:00 Quigg and C 1 (1009) N/A Straume (2000) TL seizure peak at N/A 12:10, PL seizure peak at 02:50 Herman et al. (2001) P 133 adults 57% FLE, 43% N/A (613) TLE, 40% MTLE, 24% NTLE and 13% OLE in sleep 25 S Minecan et al. R (2002) 55 adult, children (117) 61% in stage N/A S 2, 20% in stage 1, 14% in stage 3 – 4 SWS, 5% in REM Pavlova et al. R (2004) 26 adults 41% XTLE and TLE peak 15:00 – (90) 19% TLE in 19:00 (50%), XTLE sleep peak 19:00 – 23:00 S (47%) Sinha et al. R (2006) 75 adults Secondary (362) generalization N/A S MTLE peak 16:00 – I more frequently in sleep Durazzo et al. R (2008) 131 adults N/A (669) 19:00 and 07:00 – 10:00, NTLE 16:00 19:00, FLE and PLE 04:00 – 07:00, OLE 16:00 – 19:00 Hofstra et al. R (2009 a) 100 adults, Children: 36% TLE peak 05:00 – 76 children TLE and 12% 11:00 (34%) and (808) XTLE in sleep; 11:00 – 17:00 (34%) adults: 13% in children and 11:00 TLE and 30% – 17:00 in adults XTLE in sleep (37%); XTLE peak S 11:00 - 17:00 (40%) in children Hofstra et al. (2009 b) R 26 adults, 7 88% MTLE, TLE peak 11:00 – children 83% NTLE, 17:00, FLE 23:00 – (450) 83% PLE in 05:00, PLE 17:00 – wake; 100% 23:00 FLE in sleep 26 I Zarowski et al. R (2010) 100 GE more GE peak 06:00 – children frequent 12:00, FLE 03:00 – (456) during wake, 06:00 S focal epilepsy during sleep Karafin et al. R (2010) Loddenkemper R et al. (2011) 60 adults N/A MTLE peak 06:00 – (median 08:00 and 15:00 – 10/patient) 17:00 225 GE, TLE more GE peak 06:00 – children frequently 12:00, TLE 21:00 – (1008) during wake, 09:00, FLE 00:00 - PLE, FLE MLE 06:00, PLE 06:00 – in sleep, 09:00, OLE 09:00 – S S 12:00 and 15:00 – 18:00 Pavlova et al. (2012) R 44 adults N/A (129) FLE cluster around S 05:15 – 07:30, TLE cluster around 18:45 – 23:56 Table 2. Studies in humans with epilepsy analyzing the diurnal variation of the occurrence of seizures and the relationship between seizures and sleep-wake states. Focal seizures restricted to the temporal lobe tend to occur during daytime and in wakefulness. Frontal lobe and secondarily generalized seizures are more frequent during sleep. Time in 24 hour clock mode. Study design: C – case report, P – prospective, R – retrospective. Epilepsy syndromes: FLE – frontal lobe epilepsy, MTLE – mesial temporal lobe epilepsy, NTLE – neocortical temporal lobe epilepsy, GE – generalized epilepsy, FLE – frontal lobe epilepsy, PLE – parietal lobe epilepsy, OLE – occipital lobe epilepsy, TLE – temporal lobe epilepsy, XTLE – extra temporal lobe epilepsy. EEG: I – intracranial, S – surface. IEDs – interictal epileptiform discharges, SWDs – spike-wave discharges, SWS – slow-wave sleep, REM – rapid eye movement sleep, N/A – information not available. Modified after Loddenkemper et al., 2011 Childhood absence epilepsy (CAE) Childhood absence epilepsy (CAE) belongs to a group of genetic, generalized epilepsies. A positive family history is present in 14 to 45% 27 of these people and a concordance of 70 – 85% in monozygotic twins and 30% in first degree relatives (Berkovic, 1998; Bianchi et al., 1995). The genotype of the disease is complex, including autosomal-dominant gene(s) mutation(s) determining typical EEG trait and channelopathies (Crunelli and Leresche, 2002). CAE is an age-related syndrome with onset between 4 and 10, most usually between 5 and 7 years of age, and the annual incidence rate of 6.3 – 8.0/100000 (Blom et al., 1978; Olsson, 1988; Loiseau et al., 1990). It is characterized by frequent (10 to hundreds per day) short (about 10 sec.), typical absence seizures occurring awareness with and severe impairment responsiveness) in of consciousness otherwise healthy (reduced children. Clinically, absences manifests as an abrupt decrease of consciousness with cessation of ongoing voluntary activity accompanied by eyes opening, accelerated breathing and, quite frequently, by automatisms. The ictal EEG is characterized by the presence of 2.5 - 4 Hz bilateral, generalized, synchronous, symmetrical spike-wave discharges (SWDs) of sudden onset and termination (Duncan, 1997). Besides CAE, typical absences are the key symptoms in the following epilepsy syndromes recognized by the ILAE: juvenile absence epilepsy (JAE), juvenile myoclonic epilepsy (JME) and myoclonic absence epilepsy (MEA) (Commission, 1981; Panayiotopoulos 2001). Differences in the age of onset, the frequency of SWDs and a prognosis are syndrome-related (Duncan, 1997). Proper diagnosis and implementation of pharmacological treatment ensure 80% of patients becoming seizuresfree. Ethosuximide and sodium valproate are established level A medications (efficacious or effective) as an initial monotherapy for children with newly diagnosed or untreated absence seizures, according to updated ILAE evidence review of antiepileptic drug efficacy and effectiveness (Glauser et al., 2013). Medications such as carbamazepine, phenytoin, tiagabine or vigabatrin, usually prescribed to attenuate convulsive seizures, have opposite effect: precipitating or 28 even aggravating SWDs (Peeters et al., 1988; Coenen et al., 1995; Depaulis and van Luijtelaar, 2006; Bouwman et al., 2007; Glauser et al., 2013). Generalized absence seizures are neurophysiologically and pharmacologically unique. The basic underlying mechanism appears the involvement of the cortico-thalamo-cortical neuronal network. In the course of investigations, two conflicting theories about the source of the pathological oscillations emerged. Initial studies implicated the thalamus as the pacemaker of seizure activity (centrencephalic and thalamic clock theory), while other experimental work delivered evidence for the leading role of the cortex (cortical theories) (Futatsugi and Riviello Jr., 1998; Meeren et al., 2005; Avoli, 2012). Recent studies performed with the use of well characterized and validated genetic absence animal models revealed that spike-wave discharges (SWDs), the electroencephalographic hallmark of the absences, are generated in the deep layers of the somatosensory cortex (perioral area) which at the beginning of generalization process, guide the thalamus. After this initial period, the cortex and thalamus enter a bidirectional oscillatory cross-talk mode (Meeren at al., 2002; Polack et al., 2007; Lüttjohann and van Luijtelaar, 2012). Evidence is accumulating that also in humans with absence seizures the initiating side of SWDs might be cortical as well (Westmijse et al., 2009; van Luijtelaar et al., 2014). γ-aminobutyric acid (GABA) and glutamate are the most important neurotransmitters involved in the pathophysiology of absence epilepsy governing cortico-thalamo-cortical networks. The activity in these networks is modulated by cholinergic, dopaminergic and noradrenergic neurotransmission (Snead, 1995). Absence seizures are strongly related to the level of vigilance. The relationship is inversely proportional: the number of SWDs increases together with decreasing vigilance. SWDs in patients occur predominantly during drowsiness, initial stages (I and II) of slow-wave 29 sleep and during unstable transitional periods (Halasz et al., 2002). Active wakefulness and REM sleep, both characterized by high cortical desynchronization, are the least probable for the generation of SWDs. This relationship and consolidated character of human sleep, restricted on average to 8 night hours, may contribute to the circadian distribution of absence seizures in patients with SWDs (Kellaway et al., 1980, Martins da Silva et al., 1984; Horita et al., 1991). WAG/Rij rats: a well characterized and validated genetic animal model of CAE To be considered as valid, an animal model of human seizures or epilepsy should fulfill the following criteria. First, it should have sufficient “face validity” which implies similar electroencephalographic, behavioral, etiological and pharmacological characteristics to the human condition. Second, the model should have “construct validity” which means shared theoretical etiology with the disease being modeled. Third, having “predictive validity” implies that the model should correctly predict treatment effects in patients (Sarkisian, 2001; Coenen and van Luijtelaar, 2003). Animal models of epilepsy or epileptic seizures may be generally divided into two groups: models in which seizures are induced by electrical or chemical stimulation (acquired epilepsy), and genetic epilepsy models, in which animals either carry spontaneous mutations or the mutations are induced by transgenic manipulations and the seizures are expressed spontaneously (Löscher, 2002; Löscher, 2011). Chemical induction of absence seizures in previously healthy animals may be accomplished by systemic or intracerebroventricular injections of several classes of compounds such as pentylenetetrazole (PTZ) in low doses, penicillin, γ-hydroxybutyrate (GHB), 4,5,6,7- tetrahydroisoxazolo[4,5,-c ]pyridine-3-ol (THIP) or opiates (Snead, 30 1992; Avoli, 1995; Sarkisian, 2001). Among genetic models murine strains are of a particular use. There is a variety of mice strains with a well-known mutation restricted to a single gene, encoding usually different subunits of calcium channel which results in the epileptic phenotype. Lethargic (lh/lh), stargazer (stg/stg), tottering (tg/tg), leaner (tgla/tgla) or duky (du/du) mice all exhibit SWDs in the EEG. However, apart from the presence of absence seizures, they show additional severe pathological symptoms such as ataxia or motor disabilities, which make their compatibility with CAE questionable (Noebels, 2006). Opposite to single locus mutations and the concomitance of neurological abnormalities, spontaneous SWDs in rat models: Wistar Albino Glaxo from Rijswijk (WAG/Rij) and Genetic Absence Epilepsy Rats from Strasbourg (GAERS), are determined polygenetically. Additionally to the mice models, their absence epileptic phenotype is not accompanied by neurological impairments. WAG/Rij rats and GAERS are full (>30 generations brother x sister mating) inbred strains derived from Wistar stocks, sharing similar electroencephalographic, behavioral and pharmacological features. Both strains were extensively validated in a large series of behavioral, electrophysiological and pharmacological experiments and are currently world-wide used as the best described and validated genetic animal models of human absence epilepsy (Danober et al., 1998; Coenen and van Luijtelaar, 2003; Depaulis and van Luijtelaar, 2006). Although best described, they are not the only rat strains characterized by the presence of spontaneous SWDs in the EEG. Rats, especially of inbred albino strains such as Wistar or Fisher 344, but also pigmented outbred lines such as Long-Evans or Brown-Norway develop SWDs during the process of aging (Inoue et al., 1990; Willoughby and Mackenzie, 1992; Pearce et al., 2014). However, much less validation studies were done in these latter strains and therefore their predictive and constructive validity is questionable. 31 All WAG/Rij rats start showing SWDs in their EEG against a normal background activity around 2 – 3 months of age; these SWDs increase in number and duration in the course of animals maturation. Irrespectively of gender, at 6 months of age each member of the strain shows about 16 – 20 SWDs per hour lasting on average 5 sec (Coenen and van Luijtelaar, 2003). Importantly, the behavioral manifestations of absences in the rats resemble the clinical manifestation as seen in people with absence epilepsy. Animals and people become immobile during absence seizures, processing of external stimuli and execution of voluntary motor tasks are temporarily interrupted, additional mild motor signs such as facial myoclonic jerks emerges, in rats this is most clearly expressed by twitching of the vibrissae and/or eyes (van Luijtelaar and Coenen, 1986; Drinkenburg et al., 1996; Drinkenburg et al., 2003). The frequency of SWDs in rat models is between 7 – 11 Hz, starting from about 10-11 Hz at the beginning of the discharge and lowering slightly towards end (Midzianovskaia et al., 2001; Bosnyakova et al., 2007). Although bilateral and generalized, SWDs in WAG/Rij rats show a fronto-parietal domination, moreover the spike component is more pronounced at the anterior part of the cortex, while the wave has a larger amplitude at more posterior locations (Midzianovskaia et al., 2001; Meeren et al., 2002; Sitnikova and van Luijtelaar, 2007). SWDs in WAG/Rij rats occur most frequently during passive behavioral states such as light slow-wave sleep and passive wakefulness. Unstable, transitional periods between different states of vigilance also favor seizure generation, on the contrary, states characterized by extreme level of cortical synchrony (deep slow-wave sleep) or desynchronization (active wakefulness, REM sleep) are the least probable for SWDs to occur. This relationship corresponds well with the observation made in patients with absence epilepsy (Kellaway et al., 1980; Coenen et al., 1991; Drinkenburg et al., 1991; Halasz et al., 2002). The distribution of SWDs across 24 h is not uniform. 32 Absences recur daily in a rhythmic manner with an acrophase at the early hours of the dark phase and a minimum at the beginning of the light phase of the 12:12 light-dark cycle (van Luijtelaar and Coenen, 1988). The timing of the minimum of the SWD rhythm coincides with the timing of the highest amount of deep slow-wave sleep across the sleep-wake cycle of the 24 h day. Since SWDs preferentially do not occur during deep slow-wave sleep (Drinkenburg et al., 1991), the large presence of deep slow-wave sleep might prevent the occurrence of SWDs. Only the number of SWDs shows a circadian distribution, not the mean duration of episodes of SWDs. Epileptic seizures originate from local imbalances between excitation and inhibition. Convulsive epilepsy is associated with excessive excitation mediated mainly by glutamate, often in a well circumscribed part of the hippocampal circuit. Increased local cortical excitation and enhanced GABA-ergic thalamic inhibition seems to be the underlying cause of absence seizures. Indeed, not only GABA agonists increase the number of SWDs, GABA antagonists exert an opposite effect (Peeters et al., 1989; Kaminski et al., 2001). Recent work shows that GAERS absence epilepsy is due to an increased tonic GABA-ergic inhibition in the ventro-basal complex of the thalamus, while in WAG/Rij rats an enhanced local increase in cortical excitability has been found (Cope et al., 2009; Crunelli et al., 2011; Lüttjohann et al., 2012). A hyperfunction of the glutamatergic system has been found to facilitate epileptic activity regardless of the type: convulsive versus absence epilepsy and receptors involved (Peeters et al., 1990; Ramakers et al., 1991; Peeters et al., 1994). However, potentiation of two types of metabotropic glutamate receptors, mGlu1 and mGlu5 by positive allosteric modulators, has been recently described to attenuate absence seizures in WAG/Rij rats (Ngomba et al., 2011; D’Amore et al., 2013). 33 Apart from GABA and glutamate, SWDs are also modulated by catecholamine neurotransmission. Administration of the non-selective dopamine agonist apomorphine was found to reduce the number of SWDs, while treatment with the antagonist haloperidol exerted an opposite effect (Midzianovskaia et al., 2001). Attenuation of noradrenergic transmission by low doses of clonidine, an alpha-2 noradrenergic receptor agonist, caused an increase in the occurrence of absence seizures and altered activity of the cortex and the thalamus of WAG/Rij rats (Sitnikova and van Luijtelaar, 2005). Also serotonin, a neurotransmitter from the monoamine family, modulates the occurrence of absence seizures in a receptor-dependent manner. Agonist of 5-HT1A receptor (8-OH-DPAT) increased the number of SWDs, agonist of 5-HT2C receptor and antagonist of 5-HT-7 receptor decrease SWDs (Filakovszky et al., 1999; Jakus et al., 2003; Graf et al., 2004). The phenotype of WAG/Rij rats is also characterized by having depressive-like characteristics (Sarkisova and van Luijtelaar, 2011). Also in humans there is a high comorbidity between epilepsy and depression. Dopaminergic and serotonergic systems, more precisely, their strain-dependent structural and functional abnormalities are thought to contribute to the depressive-like behavior of WAG/Rij rats. In a comparison to non-epileptic control strain WAG/Rij rats show clear behavioral changes corresponding to key symptoms of human depression: anhedonia, despair and helplessness. Anhedonia was manifested by a decreased sucrose intake in the sucrose consumption test, lower preference for sucrose over tap water in the two bottle choice test, and decreased sensitivity to reward have been reported in WAG/Rij rats (Sarkisova et al., 2003). In the forced swim test (FST) used for assessment of depressive-like behavior (behavioral despair) and evaluation of anti-depressive medication efficacy, WAG/Rij rats showed a significant increase in immobility score and a decrease in 34 active behavior, interpreted as a helplessness reaction typical of humans suffering from depression. Results of the test were subsequently reversed by chronic treatment with antidepressant medication (Sarkisova et al., 2008) and by drugs that induce antiepileptogenesis (Sarkisova et al., 2010; van Luijtelaar et al., 2013). As mentioned, the rhythmic occurrence of SWDs in WAG/Rij rats might be related to endogenous cycles, such as sleep-wake and hormonal cycles. The most obvious is an inversely proportional relationship between SWDs and deep slow-wave sleep across 24 h. The acrophase of the SWDs rhythm coincides with the trough of the rhythm of deep slow-wave sleep and deprivation of sleep including deep slowwave sleep enhances the number of SWDs (Drinkenburg et al., 1995). The distribution of SWDs may be also determined by a circadian rhythm of free glucocorticoids circulating in the blood and in the brain. The highest plasma, brain and also tissue concentration of corticosterone in the rat is found 1 – 2 h before the dark phase. In the course of the day, the level of corticosterone declines, reaching a trough just before the light phase, during which the concentration is building up again (Qian et al., 2012). Both exogenously administrated corticosterone, as well as the endogenous form of the hormone secreted in response to stress, increase the number of SWDs (Schridde and van Luijtelaar, 2004; Tolmacheva et al., 2012). Glucocorticoids are thought to take part in the synchronization process between central and peripheral oscillators, therefore, it can’t be excluded that the rhythm of SWDs is driven, to some extent by cycling corticosterone. Not only might circadian, but also infradian rhythms influence the occurrence of SWDs. It has been demonstrated that the ovarian hormone progesterone, cycling with a period of 4 days in female WAG/Rij rats, increases the number of SWDs during the dark phase of the proestrus period (van 35 Luijtelaar et al., 2001), a phenomenon that has also been found in patients (van Luijtelaar et al., 2014). Aims and outline of the thesis Considering its etiology, childhood absence epilepsy is classified as a genetic type of epilepsy implicating that the onset of the disease results from a presence of specific genotype. However, during development and once developed, many other factors contribute to the occurrence of absence seizures, as found in children with CAE and in the rodent models. In general, the character of the factors contributing to the fully developed SWDs may be either endogenous or external. A bidirectional interaction is also possible: endogenous mechanisms might be modified by external influences; exogenous factors may act differently depending on the state of an internal environment. Rhythmic occurrence of SWDs in WAG/Rij rats, especially its day-to-day consistency, suggests a possible involvement of the internal time keeping mechanism in the phenomenon observed. The nature of this contribution however, has not been fully described (van Luijtelaar and Coenen, 1988). When generated endogenously, the rhythm of SWDs should persist in an environment without functional Zeitgebers. In case SWDs are imposed by the presence of at least one of them, circadian distribution of seizures should no longer be detected. A light-dark cycle, one of the strongest Zeitgebers in many living species, including rats, has a great impact on the internal clock. Sudden changes in this cycle: phase shifts, phase reversal or, the most extreme: constant conditions or photoperiods that exceed the range of entrainment, are known to disturb rhythms and their well-defined phase-relationships (Eastman and Rechtschaffen, 1982; Yamazaki et al., 2000; Nagano et al., 2003). Internal desynchronization, a result of external-internal misalignments, 36 is found to have negative consequences for health and well-being (Arendt and Marks, 1982; Knutsson, 2003). Symptoms of many medical conditions, also epileptic seizures, occur at specific time of a day (Smolensky and Peppas, 2007). WAG/Rij rats show enhanced epileptic activity during the dark phase. However, it is not clear, whether abrupt changes in the photoperiod would affect the distribution of seizures (regardless of the origin of this distribution: internal or external). Moreover, it is of potential medical significance to evaluate the effect of the phase shift on circadian physiology and seizure activity in this particular epileptic model in order to propose better hypotheses about the effects in humans. Apart from rhythmic alternation of light and darkness, the occurrence of seizures is influenced by many other factors, some of them, e.g. the sleep-wake cycle, occurring rhythmically as well (van Luijtelaar et al., 2001; Tolmacheva et al., 2012). The contribution of a momentary state of vigilance is well established: SWDs occur more often during passive wakefulness and light slow-wave sleep, while active wakefulness, deep slow-wave and REM sleep exert an inhibitory effect (Drinkenburg et al., 1991). However, questions about variability of this relationship across 24 h, as well as, about its stability in the face of different states of the circadian timing system have not been investigated before. The distribution of vigilance states across 24 h sleep-wake cycle is not uniform. Controlled by the circadian timing system, each vigilance rhythm peaks at different time of a day (Borbély, 1975; Ibuka and Kawamura, 1975). Interestingly, daily rhythmic occurrence of absence seizures in WAG/Rij rats may be clearly explained by means of a momentary state of vigilance only when deep slow-wave sleep is considered. The maximal number of SWDs seen during the dark phase and a minimum of the rhythm seen at the beginning of the light phase coincides with the lowest and the highest amount of deep slow-wave sleep, respectively. In case of the other relationships between SWDs on the one side and the remaining 37 vigilance rhythms on the other such dependencies are rather blurred or even opposite. Therefore, SWDs-state of vigilance relationship requires detailed analysis across time. Chronobiotic (aiming at adjusting the circadian timing system) properties of exogenous melatonin and its analogues are well recognized. The hormone is reported to entrain free-running circadian rhythms in constant conditions, to produce phase shifts and also to accelerate re-entrainment after such shifts. Melatonin is often recommended as a remedy for the symptoms accompanying jet lag (Srinivasan et al., 2008). Besides the circadian timing system, exogenous melatonin was found to modulate epileptic activity exerting either a convulsive or antiepileptic effect depending on the type of seizures (Lapin et al., 1998; Musshoff and Speckmann, 2003; Yildirim and Marangoz, 2006). However, studies assessing the action of this chronobiotic on absence seizures are scarce. Melatonin given in high doses to WAG/Rij rats, entrained to the regular light-dark cycle, was found to attenuate SWDs (Kldiashvili et al., 2001). However, the study focused on direct action of the compound rather than its chronobiotic properties. Therefore, it would be advantageous to evaluate the action of melatonin or melatonergic agonists on the rhythm of SWDs in WAG/Rij rats in two experimental conditions: during entrainment and after the phase shift in the photoperiod resembling jet lag. The overall aim of the current thesis is to determine the contribution of both types of factors: endogenous and external, to the occurrence of absence seizures in WAG/Rij rats. The thesis aims to answer the following questions: 1. What is the relationship between the occurrence of SWDs and other internal factors such as the state of vigilance? Is this relationship constant across 12:12 light-dark cycle? Is the relationship modified by the state of the circadian timing system: entrained versus free-running? 38 2. What is the nature of a daily rhythmic occurrence of SWDs? Is it endogenous, generated and governed by the circadian timing system or is the occurrence of SWDs masked by a presence of external factors such as a light-dark cycle? 3. How does the rhythm of SWDs entrain to the main Zeitgeber (light-dark cycle) and how does it respond to serious disturbances of the entrainment provided by this particular Zeitgeber? 4. Are external factors with documented chronobiotic properties able to influence the occurrence of SWDs and sleep-wake states, their rhythmic occurrence during entrainment and during re-entrainment? Are they able to accelerate the speed of re-entrainment of these rhythms? Chapter 1 provides introductory information to the issues addressed in the thesis. In Chapter 2 the relationship between SWDs and states of vigilance is investigated. Two approaches were compared: momentary and circadian. The former, well-established one, focused on sleep-wake states directly preceding the occurrence of SWDs, while the latter was based on correlations and cross-correlations calculated over whole 24 h and also over the light and the dark phase separately. The duration of particular sleep-wake states scored on 2 sec epoch basis, grouped subsequently into three different bin sizes, was correlated and crosscorrelated with a total time occupied by SWDs to establish temporal relationship between these variables across 24 h as well as in the light and dark period separately. In Chapter 3 the nature of the daily rhythm of the occurrence of absence seizures in WAG/Rij rats is investigated. To establish the character of the SWDs rhythm: endogenous or imposed, rats were transferred from standard laboratory conditions, in which entrainment 39 was provided by the presence of the 12:12 light-dark cycle, to an environment in which a regularly alternating level of illumination was replaced by constant dim light. Continuous recordings of the EEG and motor activity were made during 10 baseline and the last 10 experimental days of a 20 days period in constant dim light. During the first 10 days in constant dim light only motor activity was monitored to assess whether an intrinsic period of investigated rhythms was stabilized. The occurrence of SWDs, their duration, coupling with motor activity rhythm and sleep-wake states (dependent variables) were analyzed and compared between the 10 days with and the last 10 days without the presence of Zeitgebers (independent variable). Chapters 4 and 5 focus on the entrainment provided by the 12:12 light-dark cycle and consequences of temporary disturbances in the timing of this potent Zeitgeber for the occurrence of absence seizures, their duration and coupling with different sleep-wake states. In both studies WAG/Rij rats were subjected to an 8 h phase shift in the light-dark cycle which was produced by extending the dark phase from 12 to 20 h. Re-entrainment and the speed of re-entrainment to the new photoperiod was then investigated for the 10 day follow up period. Simultaneous and continuous recordings of EEG, EMG and motor activity (body movement detected by passive infrared sensor) were performed throughout the experiments which lasts 15 and 12 days in chapter 4 and 5, respectively. In Chapter 4, the process of resynchronization was undisturbed while in Chapter 5 the putative effects of agomelatine, a potent agonist of melatonin receptors and antagonist of serotonin receptors on sleep-wake parameters, SWDs and speed of re-entrainment was additionally assessed. The compound was administered, in a dose-dependent manner, starting in an acute study, followed by the study of its effects in a chronic administration (10 days) from the first day after the phase shift and continued throughout the study. 40 A general discussion, conclusions and a summary of the thesis is presented in Chapter 6. References Abe M, Herzog ED, Yamazaki S, Straume M, Tei H, Sakaki Y, Menaker M, Block GD. 2002. Circadian rhythms in isolated brain regions. J Neurosci. 22: 350-6. Achermann P, Borbély AA. 1990. Simulation of human sleep: ultradian dynamics of electroencephalographic slow-wave activity. J Biol Rhythms. 5: 141-57. Achermann P, Dijk DJ, Brunner DP, Borbély AA. 1993. A model of human sleep homeostasis based on EEG slow-wave activity: quantitative comparison of data and simulations. Brain Res Bull. 31: 97-113. Agostino PV, Golombek DA, Meck WH. 2011. Unwinding the molecular basis of interval and circadian timing. Front Integr Neurosci. 5: 64. Arendt J, Marks V. 1982. Physiological changes underlying jet lag. Br Med J (Clin Res Ed) 284:144–6. Aschoff J. 1981. A survey on biological rhythms. In Handbook of behavioral neurobiology. Volume 4. Biological rhythms. J. Aschoff (Editor). Plenum Press, New York, London. ISBN 0-306-40585-7 Atkinson HC, Waddell BJ. 1997. Circadian variation in basal plasma corticosterone and adrenocorticotropin in the rat: sexual dimorphism and changes across the estrous cycle. Endocrinology. 138: 3842-3848. Avoli M. 1995. Feline generalized penicillin epilepsy. Ital J Neurol Sci. 16: 79-82. Avoli M. 2012. Herbert H. Jasper and the Basic Mechanisms of the Epilepsies. In: Jasper's Basic Mechanisms of the Epilepsies. 4th edition. Eds. Noebels JL, Avoli M, Rogawski MA, Olsen RW, Delgado-Escueta AV. Bethesda (MD): National Center for Biotechnology Information (US). Bazil CW, Walczak TS. 1997. Effects of sleep and sleep stage on epileptic and nonepileptic seizures. Epilepsia. 38: 56-62. Berg AT, Berkovic SF, Brodie MJ, Buchhalter J, Cross JH, van Emde Boas W, Engel J, French J, Glauser TA, Mathern GW, Moshé SL, Nordli D, Plouin P, Scheffer IE. 2010. Revised terminology and concepts for organization of seizures and epilepsies: report of the ILAE Commission on Classification and Terminology, 2005-2009. Epilepsia. 51: 676-685. Berkovic SF. 1998. In Epilepsy: a Comprehensive Textbook. Eds. Engel J Jr, Pedley TA, Lippincott–Raven, Philadelphia pp 217–224. 41 Bianchi A, The Italian League Against Epilepsy Collaborative Group in Typical Absences and Related Epileptic Syndromes. 1885. Eds. Duncan JS, Panayiotopoulos CP. Churchill Livingstone, London, pp 328-337. Bingham C, Cornélissen G, Halberg F. 1993. Power of “Phase 0” chronobiologic trials at different signal-to-noise ratios and sample sizes. Chronobiologia. 20:179–190. Blask DE. 2009. Melatonin, sleep disturbance and cancer risk. Sleep Medicine Reviews 13: 257-264. Blom S, Heijbel J, Bergfors PG. 1978. Incidence of epilepsy in children: a follow-up study three years after the first seizure. Epilepsia. 19: 343-350. Borbély AA, Huston JP, Waser PG. 1975. Control of sleep states in the rat by short lightdark cycles. Brain Res. 95: 89-101. Bosnyakova D, Gabova A, Zharikova A, Gnezditski V, Kuznetsova G, van Luijtelaar G. 2007. Some peculiarities of time-frequency dynamics of spike-wave discharges in humans and rats. Clin Neurophysiol. 118: 1736-1743. Bouwman BM, Suffczynski P, Midzyanovskaya IS, Maris E, van den Broek PL, van Rijn CM. 2007. The effects of vigabatrin on spike and wave discharges in WAG/Rij rats. Epilepsy Res. 76: 34-40. Bronstein DM, Jacobs GH, Haak KA, Neitz J, Lytle LD. 1987. Action spectrum of the retinal mechanism mediating nocturnal light-induced suppression of rat pineal gland N-acetyltransferase. Brain Res. 406: 352-356. Buijs RM, Kalsbeek A. 2001. Hypothalamic integration of central and peripheral clocks. Nat Rev Neurosci. 2: 521-6. Bünning E. 1964. The Physiological Clock. Springer Verlag, Berlin. Buonomano DV, Karmarkar UR. 2002. How do we tell time? Neuroscientist. 8: 42-51. Buonomano DV. 2007. The biology of time across different scales. Nat Chem Biol. 3: 594-7. Chan S, Debono M. 2010. Replication of cortisol circadian rhythm: new advances in hydrocortisone replacement therapy. Ther Adv Endocrinol Metab. 1: 129-38. Chiazze L Jr, Brayer FT, Macisco JJ Jr, Parker MP, Duffy BJ. 1968. The length and variability of the human menstrual cycle. JAMA. 203(6):377-80. Coenen AM, Drinkenburg WH, Peeters BW, Vossen JM, van Luijtelaar EL. 1991. Absence epilepsy and the level of vigilance in rats of the WAG/Rij strain. Neurosci Biobehav Rev. 15: 259-263. 42 Coenen AM, Blezer EH, van Luijtelaar EL. 1995. Effects of the GABA-uptake inhibitor tiagabine on electroencephalogram, spike-wave discharges and behaviour of rats. Epilepsy Res. 21: 89-94. Coenen AM, Van Luijtelaar EL. 2003. Genetic animal models for absence epilepsy: a review of the WAG/Rij strain of rats. Behav Genet. 33: 635-655. Cohen MC, Rohtla KM, Lavery CE, Muller JE, Mittleman MA. 1997. Meta-analysis of the morning excess of acute myocardial infarction and sudden cardiac death. Am J Cardiol. 79: 1512-1516. Commission of Classification and Terminology of the International League Against Epilepsy. Proposal for revised clinical and electroencephalographic classification of epileptic seizures. 1981. Epilepsia. 22: 489-501. Cope DW, Di Giovanni G, Fyson SJ, Orbán G, Errington AC, Lorincz ML, Gould TM, Carter DA, Crunelli V. 2009. Enhanced tonic GABAA inhibition in typical absence epilepsy. Nat Med. 15: 1392-1398. Crespel A, Baldy-Moulinier M, Coubes P. 1998. The relationship between sleep and epilepsy in frontal and temporal lobe epilepsies: practical and physiopathologic considerations. Epilepsia. 39: 150-157. Crunelli V, Leresche N. 2002. Childhood absence epilepsy: genes, channels, neurons and networks. Nat Rev Neurosci. 3: 371-382. Crunelli V, Cope DW, Terry JR. 2011. Transition to absence seizures and the role of GABA(A) receptors. Epilepsy Res. 97: 283-289. Dai J, Swaab DF, Buijs RM. 1997. Distribution of vasopressin and vasoactive intestinal polypeptide (VIP) fibers in the human hypothalamus with special emphasis on suprachiasmatic nucleus efferent projections. J Comp Neurol. 383:397-414. D'Amore V, Santolini I, van Rijn CM, Biagioni F, Molinaro G, Prete A, Conn PJ, Lindsley CW, Zhou Y, Vinson PN, Rodriguez AL, Jones CK, Stauffer SR, Nicoletti F, van Luijtelaar G, Ngomba RT. 2013. Potentiation of mGlu5 receptors with the novel enhancer, VU0360172, reduces spontaneous absence seizures in WAG/Rij rats. Neuropharmacology. 66: 330-338. Danober L, Deransart C, Depaulis A, Vergnes M, Marescaux C. 1998. Pathophysiological mechanisms of genetic absence epilepsy in the rat. Prog Neurobiol. 55: 27-57. Depaulis A, van Luijtelaar ELJM. 2006. Genetic models of absence epilepsy in the rat. In Models of seizures and epilepsy. Eds. Pitkänen A, Schwartzkroin PA, Moshe SL. Elsevier Academic Press, Amsterdam, pp. 233-248. Drinkenburg WH, Coenen AM, Vossen JM, Van Luijtelaar EL. 1991. Spike-wave discharges and sleep-wake states in rats with absence epilepsy. Epilepsy Res. 9: 218-224. 43 Drinkenburg WH, Coenen AM, Vossen JM, van Luijtelaar EL. 1995. Sleep deprivation and spike-wave discharges in epileptic rats. Sleep. 18: 252-256. Drinkenburg WHIM, van Luijtelaar ELJM, and Coenen AML. 1996. Information processing during aberrant electroencephalographic activity in rats. In Memory and awareness in anesthesia IIIEds. Bonke B, Bovill JG, Moerman N. ASSEN: van Gorcum. pp. 186–196. Drinkenburg WH, Schuurmans ML, Coenen AM, Vossen JM, van Luijtelaar EL. 2003. Ictal stimulus processing during spike-wave discharges in genetic epileptic rats. Behav Brain Res. 143: 141-146. Duncan JS. 1997. Idiopathic generalized epilepsies with typical absences. J Neurol. 244: 403-411. Durazzo TS, Spencer SS, Duckrow RB, Novotny EJ, Spencer DD, Zaveri HP. 2008. Temporal distributions of seizure occurrence from various epileptogenic regions. Neurology. 70: 1265-1271. Dvornyk V, Vinogradova O. & Nevo E. 2003. Origin and evolution of circadian clock genes in prokaryotes. Proc. Natl Acad. Sci. USA 100, 2495–2500. Eastman C, Rechtschaffen A. 1983. Circadian temperature and wake rhythmsof rats exposed to prolonged continuous illumination. Physiol. Behav. 31: 417-427. Elliott WJ. 1998. Circadian variation in the timing of stroke onset: a meta-analysis. Stroke. 29: 992-996. Enright JT. 1965. The search for rhythmicity in biological time-series. J Theor Biol.8:426–468. Fernández JR, Hermida RC, Mojón A. 2009. Chronobiological analysis techniques. Application to blood pressure. Philos Trans A Math Phys Eng Sci. 367: 431- 445. Filakovszky J, Gerber K, Bagdy G. 1999. A serotonin-1A receptor agonist and an Nmethyl-D-aspartate receptor antagonist oppose each others effects in a genetic rat epilepsy model. Neurosci Lett. 261: 89-92. Fisher RS, van Emde Boas W, Blume W, Elger C, Genton P, Lee P, Engel J Jr. 2005. Epileptic seizures and epilepsy: definitions proposed by the International League Against Epilepsy (ILAE) and the International Bureau for Epilepsy (IBE). Epilepsia. 46: 470-472. Fraisse P. 1984. Perception and estimation of time. Annu Rev Psychol. 35:1-36. Fries P, Nikolić D, Singer W. 2007. The gamma cycle. Trends Neurosci. 30: 309-16. Futatsugi Y, Riviello JJ Jr. 1998. Mechanisms of generalized absence epilepsy. Brain Dev. 20: 75-79. 44 Gillette MU. 1991. SCN electrophysiology in vitro: rhythmic activity and endogenous clock property. In: Klein DC, Moore RY, Reppert SM (Eds.), Suprachiasmatic nucleus: the mind’s clock. Oxford Univ. Press, New York, pp: 125-143. Glauser T, Ben-Menachem E, Bourgeois B, Cnaan A, Guerreiro C, Kälviäinen R, Mattson R, French JA, Perucca E, Tomson T; ILAE Subcommission on AED Guidelines. 2013. Updated ILAE evidence review of antiepileptic drug efficacy and effectiveness as initial monotherapy for epileptic seizures and syndromes. Epilepsia. 54: 551-63. Goldman BD. 2001. Mammalian photoperiodic system: formal properties and neuroendocrine mechanisms of photoperiodic time measurement. J. Biol. Rhythms 16, 283–301. Golombek DA, Cardinali DP. 1993. Melatonin accelerates reentrainment after phase advance of the light-dark cycle in Syrian hamsters: antagonism by flumazenil. Chronobiol Int. 10:435-41. Golombek DA, Rosenstein RE. 2010. Physiology of circadian entrainment. Physiol Rev. 90: 1063-1102. Gowers W. 1885. Course of epilepsy. In: Epilepsy and Other Chronic Convulsive Diseases: Their Causes, Symptoms and Treatment. William Wood, New York, pp. 157-164. Graf M, Jakus R, Kantor S, Levay G, Bagdy G. 2004. Selective 5-HT1A and 5-HT7 antagonists decrease epileptic activity in the WAG/Rij rat model of absence epilepsy. Neurosci Lett. 359: 45-48. Halász P. 1991. Sleep, arousal and electroclinical manifestations of generalized epilepsy with spike wave pattern. Epilepsy Res. Suppl. 2: 43-48. Halász P, Terzano MG, Parrino L. 2002. Spike-wave discharge and the microstructure of sleep-wake continuum in idiopathic generalised epilepsy. Neurophysiol Clin. 32: 38-53. Halberg F, Katinas GS, Chiba Y, Garcia Sainz M, Kovats TG, Künkel H, Montalbetti N, Reinberg A, Scharf R, Simpson H. 1973. Chronobiologic glossary of the International Society for the Study of Biologic Rhythms. Int J Chronobiol. 1:31– 63. Halberg F, Tong YL, Johnson EA. 1967. Circadian system phase, an aspect of temporal morphology: procedures and illustrative examples. In: von Mayersbach H., editor. The Cellular Aspects of Biorhythms. Berlin: Springer; pp. 20–48. Hannibal J, Fahrenkrug J. 2006. Neuronal input pathways to the brain's biological clock and their functional significance. Advances in anatomy, embryology and cell biology. 182: 1-71. 45 Hastings MH, Duffield GE, Ebling FJ, Kidd A, Maywood ES, Schurov I. 1997. Non-photic signalling in the suprachiasmatic nucleus. Biol Cell. 89: 495-503. Herman ST, Walczak TS, Bazil CW. 2001. Distribution of partial seizures during the sleep–wake cycle: differences by seizure onset site. Neurology. 56: 1453-1459. Herzog ED, Schwartz WJ. 2002. A neuronal clockwork for encoding circadian time. J. Appl. Physiol. 92: 401—408. Hinton SC, Meck WH. 1997. The 'internal clocks' of circadian and interval timing. Endeavour. 21: 82-7. Hirai K, Kita M, Ohta H, Nishikawa H, Fujiwara Y, Ohkawa S, Miyamoto M. 2005. Ramelteon (TAK-375) accelerates reentrainment of circadian rhythm after a phase advance of the light-dark cycle in rats. J Biol Rhythms. 20: 27-37. Hofman MA, Zhou JN, Swaab DF. 1996. Suprachiasmatic nucleus of the human brain: an immunocytochemical and morphometric analysis. Anat Rec. 244: 552-62. Hofstra WA, Grootemarsink BE, Dieker R, van der Palen J, de Weerd AW. 2009. Temporal distribution of clinical seizures over the 24-h day: a retrospective observational study in a tertiary epilepsy clinic. Epilepsia. 50: 2019-2026. Hofstra WA, Spetgens WP, Leijten FS, van Rijen PC, Gosselaar P, van der Palen J, de Weerd AW. 2009. Diurnal rhythms in seizures detected by intracranial electrocorticographic monitoring: an observational study. Epilepsy Behav. 14: 617-621. Honma S, Kanematsu N, Katsuno Y, Honma K. 1996. Persistence of circadian oscillation while locomotor activity and plasma melatonin levels became aperiodic under prolonged continuous light in the rat. Neurosci Lett. 216: 49-52. Honma S, Shirakawa T, Katsuno Y, Namihira M, Honma K. 1998. Circadian periods of single suprachiasmatic neurons in rats. Neurosci. Lett. 250, 157–160. Horita H, Uchida E, Maekawa K. 1991. Circadian rhythm of regular spike-wave discharges in childhood absence epilepsy. Brain Dev. 13: 200-202. Hut RA, Beersma DG. 2011. Evolution of time-keeping mechanisms: early emergence and adaptation to photoperiod. Philos Trans R Soc Lond B Biol Sci. 366: 21412154. Ibuka N, Kawamura H. 1975. Loss of circadian rhythm in sleep-wakefulness cycle in the rat by suprachiasmatic nucleus lesions. Brain Res. 96: 76-81. Illnerová H, Trentini GP, Maslova L. 1989. Melatonin accelerates reentrainment of the circadian rhythm of its own production after an 8-h advance of the light-dark cycle. J Comp Physiol A. 166: 97-102. 46 Inoue M, Peeters BW, van Luijtelaar EL, Vossen JM, Coenen AM. 1990. Spontaneous occurrence of spike-wave discharges in five inbred strains of rats. Physiol Behav. 48: 199-201. Jakus R, Graf M, Juhasz G, Gerber K, Levay G, Halasz P, Bagdy G. 2003. 5-HT2C receptors inhibit and 5-HT1A receptors activate the generation of spike-wave discharges in a genetic rat model of absence epilepsy. Exp Neurol. 184: 964972. Kamiński RM, Van Rijn CM, Turski WA, Czuczwar SJ, Van Luijtelaar G. 2001. AMPA and GABA(B) receptor antagonists and their interaction in rats with a genetic form of absence epilepsy. Eur J Pharmacol. 430: 251-159. Karafin M, St Louis EK, Zimmerman MB, Sparks JD, Granner MA. 2010. Bimodal ultradian seizure periodicity in human mesial temporal lobe epilepsy. Seizure. 19: 347-351. Kellaway P, Frost Jr JD, Crawley JW. 1980. Time modulation of spike-and-wave activity in generalized epilepsy. Ann. Neurol. 8: 491-500. Kldiashvili S, Melechov M, Morenkov E, Coenen AML, van Luijtelaar G, Chepurnov SA. 2001. Effects of the sleep-wake rhythm modulating hormone melatonin on absence seizures in the EEG of rats. In Sleep-wake research in the Netherlands. Vol 12.Eds. van Bemmel AL, Beersma DGM, Hofman WF, Ruigt GSF, Vos PJE. Dutch Society for Sleep-Wake Research, Leiden, pp. 58–61. Knutsson A. 2003. Health disorders of shift workers. Occup Med (Lond). 53: 103-8. Ko CH, Takahashi JS. 2006. Molecular components of the mammalian circadian clock. Hum Mol Genet. 2:R271-7. Kotsopoulos IA, van Merode T, Kessels FG, de Krom MC, Knottnerus JA. 2002. Systematic review and meta-analysis of incidence studies of epilepsy and unprovoked seizures. Epilepsia. 43: 1402-1409. Kowanko IC, Knapp MS, Pownall R, Swannell AJ. 1982. Domiciliary self-measurement in the rheumatoid arthritis and the demonstration of circadian rhythmicity. Ann Rheum Dis. 41: 453-455. Krout KE, Kawano J, Mettenleiter TC, Loewy AD. 2002. CNS inputs to the suprachiasmatic nucleus of the rat. Neuroscience. 110: 73-92. Langdon-Down M, Brain W. 1929. Time of day in relation to convulsions in epilepsy. Lancet: 1029-1032. Lapin IP, Mirzaev SM, Ryzov IV, Oxenkrug GF. 1998. Anticonvulsant activity of melatonin against seizures induced by quinolinate, kainate, glutamate, NMDA, and pentylenetetrazole in mice. J Pineal Res. 24: 215-218. 47 LaVail MM. 1980. Circadian nature of rod outer segment disc shedding in the rat. Invest Ophthalmol Vis Sci. 19: 407-411. Lee ML, Swanson BE, de la Iglesia HO. 2009. Circadian timing of REM sleep is coupled to an oscillator within the dorsomedial suprachiasmatic nucleus. Curr Biol. 19: 848-52. Lee TM, Zucker I. 1991. Suprachiasmatic nucleus and photic entrainment of circannual rhythms in ground squirrels. J Biol Rhythms. 6: 315-30. Liu C, Weaver DR, Jin X, Shearman LP, Pieschl RL, Gribkoff VK, Reppert SM. 1997. Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron. 19:91-102. Loddenkemper T, Lockley SW, Kaleyias J, Kothare SV. 2011. Chronobiology of epilepsy: diagnostic and therapeutic implications of chrono-epileptology. J Clin Neurophysiol. 28: 146-153. Loddenkemper T, Vendrame M, Zarowski M, Gregas M, Alexopoulos AV, Wyllie E, Kothare SV. 2011. Circadian patterns of pediatric seizures. Neurology. 76: 145153. Loiseau J, Loiseau P, Guyot M, Duche B, Dartigues JF, Aublet B. 1990. Survey of seizure disorders in the French southwest. I. Incidence of epileptic syndromes. Epilepsia. 31: 391-396. Löscher W. 2002. Animal models of epilepsy for the development of antiepileptogenic and disease-modifying drugs. A comparison of the pharmacology of kindling and post-status epilepticus models of temporal lobe epilepsy. Epilepsy Res. 50: 105123. Löscher W. 2011. Critical review of current animal models of seizures and epilepsy used in the discovery and development of new antiepileptic drugs. Seizure. 20: 359368. Lüttjohann A, van Luijtelaar G. 2012. The dynamics of cortico-thalamo-cortical interactions at the transition from pre-ictal to ictal LFPs in absence epilepsy. Neurobiol Dis. 47: 49-60. Martins da Silva A, Aarts JH, Binnie CD, Laxminarayan R, Lopes da Silva FH, Meijer JW, Nagelkerke N. 1984. The circadian distribution of interictal epileptiform EEG activity. Electroencephalogr Clin Neurophysiol. 58: 1-13. Mason R, Brooks A. 1988. The electrophysiological effects of melatonin and a putative antagonist (N-acetyltryptamine) on rat suprachiasmatic neurones in vitro. Neurosci. Lett, 95: 296–301. Matell MS, Meck WH. 2000. Neuropsychological mechanisms of interval timing behavior. Bioessays. 22: 94-103. 48 Meeren HK, Pijn JP, Van Luijtelaar EL, Coenen AM, Lopes da Silva FH. 2002. Cortical focus drives widespread corticothalamic networks during spontaneous absence seizures in rats. J Neurosci. 22: 1480-1495. Meeren H, van Luijtelaar G, Lopes da Silva F, Coenen A. 2005. Evolving concepts on the pathophysiology of absence seizures: the cortical focus theory. Arch Neurol. 62: 371-376. Megdal SP, Kroenke CH, Laden F, Pukkala E, Schernhammer ES. 2005. Night work and breast cancer risk: A systematic review and meta-analysis. European Journal of Cancer 41: 2023–2032. Meijer JH, Schwartz WJ. 2003. In search of the pathways for light-induced pacemaker resetting in the suprachiasmatic nucleus. J Biol Rhythms. 18: 235-249. Midzianovskaia IS, Kuznetsova GD, Coenen AM, Spiridonov AM, van Luijtelaar EL. 2001. Electrophysiological and pharmacological characteristics of two types of spike-wave discharges in WAG/Rij rats. Brain Res. 911: 62-70. Mikkelsen JD, Vrang N. 1994. A direct pretectosuprachiasmatic projection in the rat. Neuroscience. 62: 497-505. Minecan D, Natarajan A, Marzec M, Malow B. 2002. Relationship of epileptic seizures to sleep stage and sleep depth. Sleep. 25: 899-904. Moore JG, Halberg F. 1987. Circadian rhythm of gastric acid secretion in active duodenal ulcer: chronobiological statistical characteristics and comparison of acid secretory and plasma gastrin patterns with healthy subjects and postvagotomy and pyloroplasty patients. Chronobiol Int. 4: 101-110. Moore RY, Card JP. 1994. Intergeniculate leaflet: an anatomically and functionally distinct subdivision of the lateral geniculate complex. J Comp Neurol. 344: 40330. Moore RY, Lenn NJ. 1972. A retinohypothalamic projection in the rat. J Comp Neurol. 146: 1-14. Morin LP, Allen CN. 2006. The circadian visual system, 2005. Brain Res Rev. 51: 1-60. Morin LP. 2013. Neuroanatomy of the extended circadian rhythm system. Exp Neurol. 243: 4-20. Morris C. 1992. Editor. Academic Press Dictionary of Science and Technology. San Diego: Academic Press. Musshoff U, Speckmann EJ. 2003. Diurnal actions of melatonin on epileptic activity in hippocampal slices of rats. Life Sci. 73: 2603-2610. 49 Nagano M, Adachi A, Nakahama K, Nakamura T, Tamada M, Meyer-Bernstein E, Sehgal A, Shigeyoshi Y. 2003. An abrupt shift in the day/night cycle causes desynchrony in the mammalian circadian center. J Neurosci. 23: 6141-51. Naranjo-Rodríguez EB, Prieto-Gómez B, Reyes-Vázquez C. 1991. Melatonin modifies the spontaneous multiunit activity recorded in several brain nuclei of freely behaving rats. Brain Res Bull. 27:595-600. Ngomba RT, Santolini I, Biagioni F, Molinaro G, Simonyi A, van Rijn CM, D'Amore V, Mastroiacovo F, Olivieri G, Gradini R, Ferraguti F, Battaglia G, Bruno V, Puliti A, van Luijtelaar G, Nicoletti F. 2011. Protective role for type-1 metabotropic glutamate receptors against spike and wave discharges in the WAG/Rij rat model of absence epilepsy. Neuropharmacology. 60: 1281-1291. Noebels J. 2006. Spontaneous epileptic mutation in the mouse. In Pitkanen A, Schwartzkroin PS, Moshe S (Eds). Models of seizures and epilepsy, Elsevier Academic Press, 223-232. Olsson I. 1988. Epidemiology of absence epilepsy. I. Concept and incidence. Acta Paediatr Scand. 77: 860-866. Ouyang Y, Andersson CR, Kondo T, Golden SS, Johnson CH. 1998. Resonating circadian clocks enhance fitness in cyanobacteria. Proc. Natl Acad. Sci. USA 95, 8660– 8664. Panayiotopoulos CP. 2001. Treatment of typical absence seizures and related epileptic syndromes. Paediatr Drugs. 3: 379-403. Pastore RE, Farrington SM. 1996. Measuring the difference limen for identification of order of onset for complex auditory stimuli. Percept Psychophys. 58: 510-526. Pavlova MK, Shea SA, Bromfield EB. 2004. Day/night patterns of focal seizures. Epilepsy Behav. 5: 44-49. Pavlova MK, Shea SA, Scheer FA, Bromfield EB. 2009. Is there a circadian variation of epileptiform abnormalities in idiopathic generalized epilepsy? Epilepsy Behav. 16: 461-467. Pavlova MK, Woo Lee J, Yilmaz F, Dworetzky BA. 2012. Diurnal pattern of seizures outside the hospital: is there a time of circadian vulnerability? Neurology. 78: 1488-1492. Pearce PS, Friedman D, Lafrancois JJ, Iyengar SS, Fenton AA, Maclusky NJ, Scharfman HE. 2014. Spike-wave discharges in adult Sprague-Dawley rats and their implications for animal models of temporal lobe epilepsy. Epilepsy Behav. 32: 121-131. Peeters BWMM, Spooren WPJM, van Luijtelaar ELJM, Coenen AML. 1988. The WAG/Rij model for absence epilepsy: anticonvulsant drug evaluation. Neurosci. Res. Commun. 2: 93-97. 50 Peeters BW, van Rijn CM, Vossen JM, Coenen AM. 1989. Effects of GABA-ergic agents on spontaneous non-convulsive epilepsy, EEG and behaviour, in the WAG/RIJ inbred strain of rats. Life Sci. 45: 1171-1176. Peeters BW, van Rijn CM, Vossen JM, Coenen AM. 1990. Involvement of NMDA receptors in non-convulsive epilepsy in WAG/Rij rats. Life Sci. 47: 523-529. Peeters BW, Ramakers GM, Ellenbroek BA, Vossen JM, Coenen AM. 1994. Interactions between NMDA and nonNMDA receptors in nonconvulsive epilepsy in the WAG/Rij inbred strain. Brain Res Bull. 33: 715-718. Perlow MJ, Reppert SM, Boyar RM, Klein DC. 1981. Daily rhythms in cortisol and melatonin in primate cerebrospinal fluid. Effects of constant light and dark. Neuroendocrinology. 32: 193 – 196. Perreau-Lenz S, Kalsbeek A, Garidou ML, Wortel J, van der Vliet J, van Heijningen C, Simonneaux V, Pévet P, Buijs RM. 2003. Suprachiasmatic control of melatonin synthesis in rats: inhibitory and stimulatory mechanisms. Eur J Neurosci. 17: 221-228. Perreau-Lenz S, Pévet P, Buijs RM, Kalsbeek A. 2004. The biological clock: the bodyguard of temporal homeostasis. Chronobiol Int. 21: 1-25. Petrovsky N, McNair P, Harrison LC. 1998. Diurnal rhythms of pro-inflammatory cytokines: regulation by plasma cortisol and therapeutic implications. Cytokine. 10: 307-312. Pfeffer M, Rauch A, Korf HW, von Gall C. 2012. The endogenous melatonin (MT) signal facilitates reentrainment of the circadian system to light-induced phase advances by acting upon MT2 receptors. Chronobiol Int. 29:415-429. Pitrosky B, Kirsch R, Malan A, Mocaer E, Pévet P. 1999. Organization of rat circadian rhythms during daily infusion of melatonin or S 20098, a melatonin agonist. Am J Physiol. 277:R812-R828. Pittendrigh CS. 1960. Circadian rhythms and the circadian organization of living systems. Cold Spring Harb Symp Quant Biol. 25: 159-184. Pittendrigh CS. 1981. Circadian systems: entrainment. In Handbook of Behavioral Neurobiology Vol 4, Biological Rhythms, Eds. Aschoff J, 95–124. Plenum Press, New York. Pittendrigh CS. 1993. Temporal organisation: reflections of a Darwinian clock-watcher. Annu. Rev. Physiol. 55, 17–54. Polack PO, Guillemain I, Hu E, Deransart C, Depaulis A, Charpier S. 2007. Deep layer somatosensory cortical neurons initiate spike-and-wave discharges in a genetic model of absence seizures. J Neurosci. 27: 6590-6599. 51 Pöppel E, Giedke H. 1970. Diurnal variation of time perception. Psychologische Forschung. 34: 182-198. Portaluppi F, Tiseo R, Smolensky MH, Hermida RC, Ayala DE, Fabbian F. 2012. Circadian rhythms and cardiovascular health. Sleep Med Rev. 16: 151-166. Prendergast BJ, Nelson RJ, and Zucker I. 2002. Mammalian seasonal rhythms: Behavior and neuroendocrine substrates. In Hormones, Brain and Behavior, Eds. Pfaff DW. 2, 93-156. Elsevier Science. Qian X, Droste SK, Lightman SL, Reul JM, Linthorst AC. 2012. Circadian and ultradian rhythms of free glucocorticoid hormone are highly synchronized between the blood, the subcutaneous tissue, and the brain. Endocrinology. 153: 4346-4353. Quigg M, Clayburn H, Straume M, Menaker M, Bertram III EH. 2000. Effects of circadian regulation and rest-activity state on spontaneous seizures in rat model of limbic epilepsy. Epilepsia 41: 502-509. Quigg M, Straume M, Menaker M, Bertram III EH. 1998. Temporal distribution of partial seizures: comparison of an animal model with human partial epilepsy. Ann. Neurol. 43: 748-755. Quigg M, Straume M. 2000. Dual epileptic foci in a single patient express distinct temporal patterns dependent on limbic versus nonlimbic brain location. Ann Neurol. 48: 117-120. Ramakers GM, Peeters BW, Vossen JM, Coenen AM. 1991. CNQX, a new non-NMDA receptor antagonist, reduces spike wave discharges in the WAG/Rij rat model of absence epilepsy. Epilepsy Res. 9: 127-131. Ramgopal S, Vendrame M, Shah A, Gregas M, Zarowski M, Rotenberg A, Alexopoulos AV, Wyllie E, Kothare SV, Loddenkemper T. 2012. Circadian patterns of generalized tonic-clonic evolutions in pediatric epilepsy patients. Seizure. 21: 535-539. Redman J, Armstrong S, Ng KT. 1983. Free-running activity rhythms in the rat: entrainment by melatonin. Science. 219:1089-1091. Refinetti R, Lissen GC, and Halberg F. 2007. Procedures for numerical analysis of circadian rhythms. Biol Rhythm Res. 38: 275 – 325. Reinberg A, Gervais P, Levi F, Smolensky M, Del Cerro L, Ugolini C. 1988. Circadian and circannual rhythms of allergic rhinitis: an epidemiologic study involving chronobiologic methods. J Allergy Clin Immunol. 81: 51-62. Rensing L, Meyer-Grahle U, Ruoff P. 2001. Biological timing and the clock metaphor: oscillatory and hourglass mechanisms. Chronobiol Int. 18: 329-69. 52 Reppert SM, Weaver DR, Ebisawa T. 1994. Cloning and characterization of a mammalian melatonin receptor that mediates reproductive and circadian responses. Neuron. 13: 1177-1185. Reppert SM, Weaver DR. 2002. Coordination of circadian timing in mammals. Nature. 418: 935-941. Rigas B, Torosis J, McDougall CJ, Vener KJ, Spiro HM. 1990. The circadian rhythm of biliary colic. J Clin Gastroenterol. 12: 409-414. Roenneberg T, Merrow M. 2002. Light reception: discovering the clock-eye in mammals. Curr Biol. 12: R163-165. Schernhammer ES, Laden F, Speizer FE, Willett WC, Hunter DJ, Kawachi I, Colditz GA. 2001. Rotating Night Shifts and Risk of Breast Cancer in Women Participating in the Nurses' Health Study. J Natl Cancer Inst. 98: 1563-1568. Schernhammer ES, Laden F, Speizer FE, Willett WC, Hunter DJ, Kawachi I, Fuchs CS, Colditz GA. 2003. Night-shift work and risk of colorectal cancer in the nurses' health study. J Natl Cancer Inst. 95: 825-828. Sharkey KM, Eastman CI. 2002. Melatonin phase shifts human circadian rhythms in a placebo-controlled simulated night-work study. Am J Physiol Regul Integr Comp Physiol. 282: R454-463. Sarkisian MR. 2001. Overview of the Current Animal Models for Human Seizure and Epileptic Disorders. Epilepsy Behav. 2: 201-216. Sarkisova KY, Midzianovskaia IS, Kulikov MA. 2003. Depressive-like behavioral alterations and c-fos expression in the dopaminergic brain regions in WAG/Rij rats with genetic absence epilepsy. Behav Brain Res. 144: 211-226. Sarkisova KY, Kulikov MA, Midzyanovskaya IS, Folomkina AA. 2008. Dopaminedependent nature of depression-like behavior in WAG/Rij rats with genetic absence epilepsy. Neurosci Behav Physiol. 38: 119-128. Sarkisova KY, Kuznetsova GD, Kulikov MA, van Luijtelaar G. 2010. Spike-wave discharges are necessary for the expression of behavioral depression-like symptoms. Epilepsia. 51: 146-160. Sarkisova K, van Luijtelaar G. 2011. The WAG/Rij strain: a genetic animal model of absence epilepsy with comorbidity of depression [corrected]. Prog Neuropsychopharmacol Biol Psychiatry. 35: 854-876. Schridde U, van Luijtelaar G. 2004. Corticosterone increases spike-wave discharges in a dose- and time-dependent manner in WAG/Rij rats. Pharmacol Biochem Behav. 78: 369-375. 53 Sharma VK, Singaravel M, Subbaraj R, Chandrashekaran MK. 1999. Timely administration of melatonin accelerates reentrainment to phase-shifted lightdark cycles in the field mouse Mus booduga. Chronobiol Int. 16:163-70. Shechter A, Boivin DB. 2010. Sleep, Hormones, and Circadian Rhythms throughout the Menstrual Cycle in Healthy Women and Women with Premenstrual Dysphoric Disorder. Int Journal of Endocrinology. 2010: 259345. Shorvon SD. 2011. The etiologic classification of epilepsy. Epilepsia. 52: 1052-1057. Sinha S, Brady M, Scott CA, Walker MC. 2006. Do seizures in patients with refractory epilepsy vary between wakefulness and sleep? J Neurol Neurosurg Psychiatry. 77: 1076-1078. Sitnikova E, van Luijtelaar G. 2005. Reduction of adrenergic neurotransmission with clonidine aggravates spike-wave seizures and alters activity in the cortex and the thalamus in WAG/Rij rats. Brain Res Bull. 64: 533-540. Sitnikova E, van Luijtelaar G. 2007. Electroencephalographic characterization of spikewave discharges in cortex and thalamus in WAG/Rij rats. Epilepsia. 48: 22963311. Slotten HA, Pitrosky B, Pévet P. 1999. Influence of the mode of daily melatonin administration on entrainment of rat circadian rhythms. J Biol Rhythms. 14:347-353. Smolensky MH, Peppas NA. 2007. Chronobiology, chronotherapeutics. Adv Drug Deliv Rev. 59: 828-51. drug delivery, and Smolensky MH, Reinberg A, Labrecque G. 1995. Twenty-four hour pattern in symptom intensity of viral and allergic rhinitis: treatment implications. J Allergy Clin Immunol. 95: 1084-1096. Snead OC 3rd. 1992. Pharmacological models of generalized absence seizures in rodents. J Neural Transm Suppl. 35:7-19. Snead OC 3rd. 1995. Basic mechanisms of generalized absence seizures. Ann Neurol. 37: 146-157. Snyder F, Hobson JA, Morrison DF, Goldfrank F. 1964. Changes in respiration, heart rate, and systolic blood pressure in human sleep. J Appl Physiol. 19: 417-22. Solomon GD. 1992. Circadian rhythms and migraine. Cleve Clin J Med. 59: 326-329. Spadoni G, Bedini A, Rivara S, Mor M. 2011. Melatonin receptor agonists: new options for insomnia and depression treatment. CNS Neurosci Ther. 17: 733-741. Srinivasan V, Singh J, Pandi-Perumal SR, Brown GM, Spence DW, Cardinali DP. 2010. Jet lag, circadian rhythm sleep disturbances, and depression: the role of melatonin and its analogs. Adv Ther. 27: 796-813. 54 Srinivasan V, Spence DW, Pandi-Perumal SR, Trakht I, Cardinali DP. 2008. Jet lag: therapeutic use of melatonin and possible application of melatonin analogs. Travel Med Infect Dis. 6: 17-28. Stankov B, Fraschini F, Reiter RJ. 1991. Melatonin binding sites in the central nervous system.Brain Res Brain Res Rev. 16: 245-256. Tapia-Osorio A, Salgado-Delgado R, Angeles-Castellanos M, Escobar C. 2013. Disruption of circadian rhythms due to chronic constant light leads to depressive and anxiety-like behaviors in the rat. Behav Brain Res. 252: 1-9. Tolmacheva EA, Oitzl MS, van Luijtelaar G. 2012. Stress, glucocorticoids and absences in a genetic epilepsy model. Horm Behav. 61: 706-710. Vanecek J.1998. Cellular mechanisms of melatonin action. Physiol Rev. 78: 687-721. van Luijtelaar EL, Coenen AM. 1986. Two types of electrocortical paroxysms in an inbred strain of rats. Neurosci Lett. 70: 393-397. van Luijtelaar EL, Coenen AM. 1988. Circadian rhythmicity in absence epilepsy in rats. Epilepsy Res. 2: 331-336. van Luijtelaar G, Budziszewska B, Jaworska-Feil L, Ellis J, Coenen A, Lasoń W. 2001. The ovarian hormones and absence epilepsy: a long-term EEG study and pharmacological effects in a genetic absence epilepsy model. Epilepsy Res. 46: 225-239. van Luijtelaar G, Mishra AM, Edelbroek P, Coman D, Frankenmolen N, Schaapsmeerders P, Covolato G, Danielson N, Niermann H, Janeczko K, Kiemeneij A, Burinov J, Bashyal C, Coquillette M, Lüttjohann A, Hyder F, Blumenfeld H, van Rijn CM. 2013. Anti-epileptogenesis: Electrophysiology, diffusion tensor imaging and behavior in a genetic absence model. Neurobiol Dis. 60: 126-138. van Luijtelaar G, Behr C, Avoli M. 2014. Is there such a thing as "generalized" epilepsy? Adv Exp Med Biol. 813: 81-91. van Luijtelaar G, Onat FY, Gallagher MJ . 2014. Animal models of absence epilepsies: what do they model and do sex and sex hormones matter? Neurobiol Dis. 72: 167-179. von Steinbüchel N. 1995. Temporal system states in speech processing. In: Supercomputing in brain research: from tomography to neural networks. Eds. Herrmann HJ, Wolf DE, Pöppel E. World Scientific, Singapore, 75-81. Waterhouse J, Drust B, Weinert D, Edwards B, Gregson W, Atkinson G, Kao S, Aizawa S, Reilly T. 2005. The circadian rhythm of core temperature: origin and some implications for exercise performance. Chronobiol Int. 22: 207-25. 55 Weaver DR, Rivkees SA, Reppert SM. 1989. Localization and characterization of melatonin receptors in rodent brain by in vitro autoradiography. J Neurosci. 9: 2581-2590. Wehr TA. 1982. Circadian rhythm disturbances in depression and mania, in: Rhythmic Aspects of Behavior, Eds. Brown FM, Graeber RC. Lawrence Erlbaum Ass., New Jersey, 399–428. Welsh DK, Logothetis DE, Meister M, Reppert SM. 1995. Individual neurons dissociated from rat suprachiasmatic nucleus express independently phased circadian firing rhythms. Neuron 14, 697–706. Westmijse I, Ossenblok P, Gunning B, van Luijtelaar G. 2009. Onset and propagation of spike and slow wave discharges in human absence epilepsy: A MEG study. Epilepsia. 50: 2538-2548. Willoughby JO, Mackenzie L. 1992. Nonconvulsive electrocorticographic paroxysms (absence epilepsy) in rat strains. Lab Anim Sci. 42: 551-554. Wittmann M. 1991. Time perception and temporal processing levels of the brain. Chronobiol Int. 16: 17-32. Woelfle MA, Ouyang Y, Phanvijhitsiri K, Johnson CH. 2004. The adaptive value of circadian clocks: an experimental assessment in cyanobacteria. Curr. Biol. 14, 1481–1486. Woodfill CJ, Wayne NL, Moenter SM, Karsch FJ. 1994. Photoperiodic synchronization of a circannual reproductive rhythm in sheep: identification of season-specific time cues. Biol Reprod. 50: 965-976. Yamazaki S, Numano R, Abe M, Hida A, Takahashi R, Ueda M, Block GD, Sakaki Y, Menaker M, Tei H. 2000. Resetting central and peripheral circadian oscillators in transgenic rats. Science. 288: 682-685. Yildirim M, Marangoz C. 2006. Anticonvulsant effects of melatonin on penicillin-induced epileptiform activity in rats. Brain Res. 1099: 183-188. Zarowski M, Loddenkemper T, Vendrame M, et al. 2010. Relationship of day/night and sleep/wake patterns to seizure semiology and epilepsy localization. Neurology. 74 (suppl 2): A105 (abstract). Zordan MA, Rosato E, Piccin A, Foster R. 2001. Photic entrainment of the circadian clock: from Drosophila to mammals. Semin Cell Dev Biol. 12: 317-28. 56 CHAPTER 2 Differential temporal organization of sleep-wake states and absence seizures in light versus dark phase in a genetic animal model Submitted to Epilepsy Research Abstract The goal of the present study was to investigate how spikewave discharges and different sleep-wake states are temporally related. Continuous electroencephalographic (EEG) and electromyographic (EMG) recordings across 24 h were made in adult male WAG/Rij rats kept in a 12:12 light-dark cycle (LD 12:12). Three different resolutions of the data: 1 h, 6 min and 0.5 min were used. Total time of a particular state of vigilance and total time occupied by spike-wave discharges was correlated and cross-correlated over 24 h and for the light and dark phase separately. Strong positive correlation between spike-wave discharges and active wakefulness, and strong negative ones with light and deep slow-wave sleep for 1 h bin size were found. Correlations were lower with the increase in resolution. Large phase-related differences in the temporal organization of spike-wave discharges and sleep-wake states were seen: unidirectional in the light and bidirectional in the dark phase. It was thought that this is most likely due to the entraining power of the light onset. Keywords: vigilance/sleep-wake absence state, epilepsy, light and WAG/Rij dark rats, phase, state of spike-wave discharges, circadian rhythm 1. Introduction Childhood Absence Epilepsy (CAE) is an epileptic syndrome of generalized, nonconvulsive seizures, which are manifested as short moments of decreased responsiveness coinciding with 2 – 4 Hz SpikeWave Discharges (SWDs) in the EEG (Duncan, 1997; Engel Jr, 2006; Niedermeyer, 1972). In WAG/Rij rats, an often used genetic absence epilepsy rodent model, 7 – 11 Hz SWDs are considered as the EEG 59 equivalents of absence seizures (Depaulis and van Luijtelaar, 2006; Sitnikova and van Luijtelaar, 2007). During seizures animals are immobile and unresponsive; pharmacological therapeutics against absence epilepsy decrease the number and duration of SWDs (Depaulis and van Luijtelaar, 2006). Absence seizures in patients occur typically during drowsiness, non-rapid-eye movement sleep (non-REM sleep, stages I and II) and also around transitions between sleep-wake states (Halasz et al., 2002). Similarly, in WAG/Rij rats, SWDs are preceded mostly by passive wakefulness and light slow-wave sleep. More specifically, analysis of short lasting (5 s) EEG epochs directly preceding and following SWDs (this is called the momentary approach) has confirmed that animals have seizures mostly during unstable, transitional periods across the sleep-wake cycle. A highly desynchronized EEG typical for active wakefulness or rapid-eye movement sleep (REM), as well as highly synchronized one, typical for deep slow-wave sleep, are not a favorable temporal context for the occurrence of SWDs (Drinkenburg et al., 1991; Lüttjohann et al., 2012). This implies a large negative relation between the occurrence of active wakefulness and deep slowwave sleep on the one hand and SWDs on the other. A 24 h rhythm in the occurrence of SWDs has been detected under entrainment to the LD 12:12: absences reach a maximum during the dark phase, while they are almost absent during the first few hours of the light phase (Smyk et al., 2011; Smyk et al., 2012; van Luijtelaar and Coenen, 1988). A 24 h periodicity is also observed for the timing of the different states of vigilance, which taken together, constitute the sleep-wake cycle (Borbély et al., 1975; Daan et al., 1984). The circadian rhythm and momentary factors controlling the occurrence of SWDs interact: this is illustrated during the timing of the minimum of the rhythm. The beginning of the light phase, when SWDs rarely occur, is the period with the highest amount of deep slow-wave sleep 60 (Borbély, 1975; Drinkenburg et al., 1991; Friedman et al., 1979; van Luijtelaar and Coenen, 1984). However, active wakefulness, which did not precede SWDs when momentary factors were taking into consideration, predominates and peaks during the dark period. This demonstrates that active wakefulness and SWDs coincide on the 24 h scale but dissociate when momentary factors are taking into account. The goal of the present study is to (re)investigate the temporal relationship between SWDs and the different sleep-wake states across the 24 h cycle by means of correlation and cross correlation functions. These methods were applied on 24 h EEG data at different resolutions (1 h, 6 min and 30 s) which together could capture both: a description and temporal organization on a resolution commonly used in circadian research and on a much shorter time scale, suited for considering momentary factors determining the occurrence of SWD. This was done also separately for the light and dark phase. 2. Materials and methods 2.1. Animals Twelve adult, male WAG/Rij rats (Harlan, the Netherlands) weighting 315 g ± 12g (eight months of age) were used. Animals were singly-housed in individually ventilated Plexiglas cages (25 x 22 x 18 cm) in a sound-attenuated room and controlled environmental conditions (temperature: 22 ± 2 oC, humidity: 60%, 12:12 light-dark cycle, white lights on at 05:00 am off at 17:00 pm, light intensity: ≈ 100 lux) throughout the experiment as well as during the recovery period after the surgery. Standard laboratory chow and tap water were available ad libitum. All experimental procedures were approved by the animal use and care committee of Janssen Research and Development, 61 Pharmaceutical Companies of Johnson & Johnson and the local ethical committee. 2.2. Surgery Under deep isoflurane anesthesia seven stainless steel fixing screws (diameter 1 mm) were inserted bilaterally in the left and right hemisphere along the antero-posterior axes in the following coordinates: AP +2 mm, L +/– 2 mm; AP -2 mm, L +/-2 mm and AP 6.6 mm, L +/- 2 mm from Bregma; and referenced to the same ground electrode place midline above the cerebellum. The incisor bar was –5 mm under the center of the ear bar (Paxinos and Watson, 1998). In addition, stainless steel wire EMG electrodes were placed in the muscles of the neck. Electrodes (stainless steel wire, 7N51465T5TLT, 51/46 Teflon Bilaney, Germany) were connected to a pin (Future Electronics: 0672-2-15-15-30-27-10-0) with a small insert (track pins; Dataflex: TRP-1558-0000) and were fit into a 10-hole connector, then the whole assembly was fixed with dental cement to the cranium. Rats were allowed to recover for 10 days after surgery. 2.3. EEG and EMG recordings Rats, while staying in their home cages, were placed in the sound-attenuated, recording boxes and connected by a cable to rotating swivels, allowing free movement during recording procedures. Rats were allowed to habituate to the recording set-up for 24 h preceding the experiment. Continuous EEG and EMG signals were acquired at a 2 kHz sample rate with an input range of +/- 500 mV through a Biosemi ActiveTwo system (Biosemi, Amsterdam, the Netherlands) referenced to the CMS-DRL ground (common mode reference for online data acquisition and impedance measures, which 62 is a feedback loop driving the average potential across the montage close to the amplifier zero). The signals were amplified, band-pass filtered between 1 and 100 Hz, and digitized with 24 bit resolution. 2.4. Experimental design Data used in the present study were from a larger study in which the putative action of agomelatine, a newly introduced antidepressant agent with chronobiological properties, on SWDs and sleep-wake states, was investigated (Chapter 5). Twenty four hours EEG and EMG recordings were performed during 3 consecutive days. On the first day, animals received a peri oral (p.o.) injection of saline 1 hour before the onset of darkness. Only data from the first day (24 h of EEG and EMG recordings in LD 12:12 with p.o. saline injection 1 h before the onset of darkness) were analyzed here. 2.5. Data analysis 5 vigilance states: active wakefulness, passive wakefulness, light and deep slow-wave sleep, REM sleep, as well as a number of transitions between them, were identified automatically for 2 s epochs based on EEG, EMG and body movements (passive infrared, PIR, detection) following validated criteria (Ahnaou and Drinkenburg, 2011). The occurrence and duration of SWDs across 24 h was scored automatically based on the EEG signal from a frontal electrode according to well-established criteria, next they were verified visually (Ovchinnikov et al., 2010; van Luijtelaar and Coenen, 1986). Data from 24 h: time spent in a particular state of vigilance (expressed in s) and total duration of SWDs (also expressed in s) were subsequently analyzed with 3 different bin sizes: 1 h, as is commonly 63 done in chronobiology, 6 min and 30 s, allowing a sufficient resolution considering the duration of the sleep cycle, non-REM and REM duration and the clustering of SWDs in a minute range in WAG/Rij rats (Midzianovskaya et al., 2006; van Luijtelaar and Bikbaev, 2007). Pearson’s correlations and cross-correlations were calculated to estimate relationships between total duration of SWDs on the one side, and time spent in a particular sleep-wake state and transitions between different states of vigilance on the other across the 24 h period for each individual subject. Cross-correlations for 1 h resolution (bin size) of the data were calculated for 24 time lags, while for 6 min and 0.5 min resolutions of 60 time lags were analyzed. These calculations were made for the 24 h data, as well as for the 12 h light and 12 h dark phase, respectively. Autocorrelation and spectral density (single spectrum Fourier analysis) of individual data of total duration of SWDs and sleep-wake states in the light and dark period separately (resolution of the data: 6 min) were used to analyze the temporal organization and rhythmicity of SWDs and sleep-wake states separately. 2.6. Statistical analysis The statistical analyses were done with Statistica (StatSoft Inc., Tulsa, Oklahoma, USA). Pearson’s correlation coefficients were used to correlate across time (hours) the relationships between the various vigilance states and SWDs (R, p < 0.05) and the mean and SEM of the correlation coefficients across 12 animals were calculated, and confidence limits (> than 2 times their respective standard errors) were used, the differences between light and dark were tested with Student t-test, p < 0.05. 64 3. Results 3.1. Pearson’s correlations for 24 hours The distribution of the sleep-wake states and SWDs over the 24 h period in the 1 h resolution bin size is presented in Figure 1 A. The rhythms of active wakefulness and SWDs were positively correlated over the 24 h period (R = 0.75 ± 0.04, p < 0.05), and those of deep and light slow wave sleep and SWDs were negatively correlated over time (R = -0.70 ± 0.05, p < 0.05 and R = -0.55 ± 0.05, p < 0.05, respectively: Fig. 1 C). Pearson’s correlation coefficients became small and insignificant for the 6 and 0.5 min bins. The total time of SWDs was positively correlated with transitions from active to passive wakefulness and from passive to active wakefulness (R = 0.59 ± 0.10, p < 0.05 and R = 0.59 ± 0.10, p < 0.05, respectively), and negatively correlated with transitions from light to deep slow-wave sleep and from deep to light slow-wave sleep (R = -0.63 ± 0.07, p < 0.05 and R = 0.62 ± 0.07, p < 0.05, respectively; a 24 h plot of the number of transitions and SWDs is presented in Figure 1 B). The decrease of the bin size from 1h to 6 and 0.5 min was accompanied with a decrease of the values of Pearson’s correlation coefficients, which became insignificant. 65 Figure 1. A) Mean of total time of sleep-wake states and total time of SWDs across 24 h in a resolution of 1 h (upper graph) and 6 min (lower graph) (n = 12). AW – orange line – active wakefulness, PW – red line - passive wakefulness, SWS L – light green line - light slow-wave sleep, SWS D – dark green line - deep slow-wave sleep, REM – blue line - REM sleep. The dark phase of the LD 12:12 is marked by shaded background. B) Mean of the number of transitions between sleep-wake states across 24 h in a resolution of 1 h. AW – PW – transitions from active to passive wakefulness, PW – AW – transitions from passive to active wakefulness, SWS L – SWS D – transitions from light to deep slow-wave sleep, SWS D – SWS L – transitions from deep to light slowwave sleep. The dark phase of the LD 12:12 is marked by grey rectangle. C) Pearson’s correlation coefficients (mean ± SEM) obtained for the total time of sleep-wake states and the total time of SWDs for 1 h, 6 and 0.5 min resolution of the data. * p < 0.05 according to the critical value table for Pearson’s correlation coefficients 66 3.2. Cross-correlations for 24 hours Significant cross-correlations were found between SWDs on the one side and active wakefulness, light and deep slow-wave sleep on the other for the 1 h resolution data (Figure 2 A). The occurrence of the maximum at lag 0 for active wakefulness demonstrates the cooccurrence of active wakefulness with SWDs. The negative value of the maximum at lag 0 for deep slow-wave sleep suggests that SWDs and deep slow-wave sleep do not co-occur. The highest cross-correlation coefficient for light slow-wave sleep, seen at lag -2 (-4 to -1), suggests that the duration of SWDs may predict the duration of light slow-wave sleep in the consecutive 1 to 4 hours. SWDs and sleep-wake states were best cross-correlated at positive time lag 1 for the 6 min resolution of the data; only for REM sleep none of the cross-correlation coefficients exceeded the confidence limits (Figure 2 B). Apart from time lag 1, small significant cross-correlations were also observed at other positive, as well as negative time lags. This suggests a bidirectional relation between SWDs and these vigilance states. Not only may the duration of particular sleep-wake states predict a higher probability of an increase of the time occupied by SWDs, but also the time occupied by SWDs influences the amount of vigilance states in the consecutive minutes. Data are presented in Figure 2 B. Significant cross-correlation coefficients between SWDs and sleep-wake states expressed in 0.5 min resolution were seen in all states predominantly with positive time lags (Figure 2 C). REM sleep was an exception: it was significantly cross-correlated with SWDs almost only at negative time lags. Some positive cross-correlation coefficients were observed for active wakefulness and passive wakefulness and lower cross-correlations and less time lags for passive wakefulness, negative for light and deep slow-wave sleep and REM sleep. Two features of cross-correlation coefficients in the 0.5 min 67 resolution of the data are striking. First, they were relatively small. Second, there was no single, sharp peak in a range of significant crosscorrelation coefficients. Instead, there was a large number (up to 50 60, 25 – 30 min) of significant time lags, which, together with their small values suggest that the strength of the temporal relationship between SWDs and these states is small, stable and that the occurrence of SWDs is determined by the vigilance states over a period of 25 to 30 min. 68 Figure 2. Cross-correlations of total time of sleep-wake states and total time of SWDs for 1 h (A), 6 min (B) and 0.5 min resolution of the data (C) (n = 12). AW – orange line - active wakefulness, PW – red line - passive wakefulness, SWS L – light green line - light slow-wave sleep, SWS D – dark green line 69 deep slow-wave sleep, REM – blue line - REM sleep. Cross-correlation coefficients are given in mean ± SEM. The time lag 0 is marked by the black dashed line, confidence limits (> than 2 times respective standard errors) in red dashed line 3.3. Pearson’s correlation for the dark and the light phase The strength of the temporal relationship between SWDs and sleep-wake states, as expressed by the correlation coefficients, was almost always stronger in the light than in the dark phase, irrespectively of the bin size. Correlation coefficients for SWDs and active wakefulness in 1 h and 6 min resolution were both positive and larger in the light than in the dark phase for 1 h (0.64 ± 0.04 vs. 0.40 ± 0.07, p < 0.05) and for the 6 min bin size (0.30 ± 0.09 vs. 0.05 ± 0.08, p < 0.05). Also in the 0.5 min resolution stronger temporal relationship was found for the light than for the dark phase (-0.13 ± 0.01 vs. 0.06 ± 0.01, p < 0.05). Similar differences between light and dark were found for the relationship between SWDs and passive wakefulness, again irrespective of bin size (all p’s < 0.05) and as well as between SWDs and deep slow wave sleep. Deep slow-wave sleep was always negatively correlated with SWDs. This correlation coefficient was also always higher in the light than in the dark phase (-0.69 ± 0.08 vs 0.39 ± 0.19, p < 0.05 for 1 h; -0.33 ± 0.04 vs 0.10 ± 0.11, p < 0.05 for 6 min and - 0.14 ± 0.01 vs. -0.02 ± 0.01, p < 0.05 for 0.5 min resolution of the data). The correlation between SWDs and REM sleep was higher and negative in the dark and positive and lower in the light phase irrespective of bin size, however, none of the correlation coefficients reached significance. 70 3.4. Cross-correlations during the dark and the light phase The 1 h resolution of the data showed clear peaks for crosscorrelations of SWDs with active wakefulness, deep slow-wave sleep and REM sleep at the time lag 0 in the dark period, nonetheless they did not exceed confidence limits (Figure 3 A). In contrast, sharp peaks at the time lag 0 for active wakefulness, deep slow-wave sleep and passive wakefulness were seen in the light phase, these for active wakefulness and deep slow wave sleep were significant (Figure 3 B). Clear light-dark related differences were also seen in the other two bin sizes. The direction of cross-correlations in the light phase for 6 and 0.5 min resolutions (Figure 4 A and B, respectively) was the same as obtained for 1h: positive for active and passive wakefulness, negative for light and deep slow-wave sleep. However, in the dark phase, next to being significant at the same lag and in the same direction as those observed for the light phase, an additional peak(s) with significant cross-correlations for active wakefulness, light and deep slow-wave sleep emerged. These additional peaks were observed at negative time lag -1 (cross-correlation’s value -0.2 ± 0.06 for active wakefulness, 0.28 ± 0.05 for light slow-wave sleep and 0.21 ± 0.08 for deep slowwave sleep in 6 min resolution of the data) and these cross-correlations were negative. In the highest resolution of the data (0.5 min) these additional peaks were again observed in the dark phase. However, these peaks were not sharp. Instead, more than one cross-correlation coefficient exceeded confidence limits, and their values were smaller in comparison to 6 min resolution. Apart from a characteristic decrease in the value of crosscorrelation coefficients across the data resolution, which might suggest that the strength of the relationship between SWDs and sleep wake states becomes smaller with a smaller bin size, clear differences 71 between phases were noticed. In the light phase, behavior of particular sleep-wake state might be seen as a predictor of the occurrence of SWDs. Presence of additional significant cross-correlations at negative time lags in the dark phase suggests that the relationship between SWDs and vigilance states in this phase is bidirectional. Not only can sleep-wake states predict the probability of the occurrence of SWDs but also the occurrence of SWDs may predict the probability of the occurrence of consecutive sleep-wake states. Figure 3. Cross-correlations of total time of sleep-wake states and total time of SWDs for 1 h resolution of the data in the dark (A) and the light phase (B) 72 (n = 12). AW – orange line – active wakefulness, PW – red line - passive wakefulness, SWS L – light green line - light slow-wave sleep, SWS D – dark green line - deep slow-wave sleep, REM – blue line - REM sleep. Crosscorrelation coefficients are given in mean and SEM. The time lag 0 is marked by the black dashed line, confidence limits (> than 2 times respective standard errors) in red dashed line 3.5. Autocorrelations and spectral density for the dark and the light phase Clear light-dark differences were found for both autocorrelation and spectral density functions. Significant autocorrelation coefficients were found mainly for the first (SWDs and passive wakefulness), first and second (light slow-wave sleep) and 1st to 5th time lags (deep slowwave sleep) in the light phase, while during the dark phase autocorrelations were either insignificant (SWDs, passive wakefulness) or restricted to the first time lag (light and deep slow-wave sleep). There were no differences between the dark and the light phase for active wakefulness and REM sleep. In general, higher spectral density, for practically all periods, was observed in the dark phase for SWDs, active wakefulness, and REM sleep. Time series of SWDs were characterized by high spectral density in a range of short periodicities (12 to 42 min) with two highest peaks at 42 min and also in 6 h in the dark phase. In the light phase, spectral density was lower, with maxima at 1.5 and 2.4 h. Similarly, active wakefulness showed higher spectral density in the dark phase with three dominant peaks at 48 min, 1 h and 1.5 h, while in the light phase spectral density was lower, peaking at 42 min, 1.2 and 1.7 h. Dominant periodicities in REM sleep in the dark phase were: 48 min, 1.5 and 4 h; in the light phase: 24 min, 54 min and 1.7 h. Spectral density in the dark phase was found to be higher in comparison to the light phase. Contrary to SWDs and sleep-wake states mentioned above, passive wakefulness, light and deep slow-wave sleep showed greater values of spectral density in the 73 light phase. The occurrence of passive wakefulness in the dark phase was characterized by many short periodicities within a range of 12 min to 1.3 h with a low spectral density in comparison to the light phase with one dominant peak at 1.7 h. Similarly, light slow-wave sleep showed higher spectral density in the light phase (dominant peaks: 1.2; 1.7 and 6 h), in the dark phase those peaks were shifted towards shorter periodicities, implicating higher frequencies. The occurrence of deep slow-wave sleep in the dark period was dominated by three periodicities: 48 min, 1 and 1.3 h. In the light phase, apart from greater values of spectral density, a shift in maximal value (1.7 h and 6 h) was observed. An example of autocorrelation and spectral density functions of SWDs and deep slow-wave sleep of a single animal in the dark and light period is presented in Figure 5. 74 75 Figure 4. Cross-correlations of total time of sleep-wake states and total time of SWDs in the light and the dark phase for 6 min (A) and 0.5 min (B) resolution of the data (n = 12). AW – orange line - active wakefulness, PW – red line passive wakefulness, SWS L – light green line - light slow-wave sleep, SWS D – dark green line - deep slow-wave sleep, REM – blue line - REM sleep. Crosscorrelation coefficients are given in mean and SEM. The time lag 0 is marked by the black dashed line, confidence limits (> than 2 times respective standard errors) in red dashed line Figure 5. An example of autocorrelation and spectral density functions of SWDs (A and C, respectively) and deep slow-wave sleep (B and D, respectively) in the light and the dark phase for 6 min resolution of the data of a single subject. Confidence limits (> than 2 times respective standard errors) for the autocorrelations are marked by the red dashed lines 76 4. Discussion The temporal relationship between SWDs and sleep-wake states was investigated by means of correlation and cross-correlation functions applied across the 24 h period with different resolutions. Firstly, strong correlations were found between the total time of SWDs and active wakefulness, and deep slow-wave sleep for the 1 h resolution data. Secondly, detailed cross-correlation analysis of higher resolutions revealed that SWDs and sleep-wake states predict each other’s occurrence. Thirdly, the strength of the temporal relationship between SWDs and sleep-wake states was different between the light and the dark phase. A relationship between the occurrence of absence seizures and vigilance states is commonly investigated by means of the momentary approach, the identification of sleep-wake state directly preceding SWDs (Drinkenburg et al., 1991; Lannes et al. 1988). Based on previously established relationships (Drinkenburg et al., 1991), one might predict that the direction of both correlations: active wakefulness - SWDs and deep slow-wave sleep – SWDs, should be negative. However, active wakefulness appeared to be strongly and positively correlated with SWDs at the 1 h bin, this means that both functions cooccur over time. Although a bin length of 1 h is a relatively large time scale in a view of the dynamics of rat sleep-wake organization, it reveals a close relationship between SWDs and active wakefulness. Their highly daily rhythmic occurrence of active wakefulness/motor activity and absence seizures suggest an influence of the circadian timing system (Ibuka et al., 1975; Smyk et al., 2011; Stephan and Zucker, 1972). Under LD 12:12, the shape of these rhythms is similar, as could be seen in Fig. 1. Both functions dominate during the dark phase, while their minimal level is seen during the first few hours of the light phase (Smyk et al., 2011, Smyk et al., 2012; van Luijtelaar 77 and Coenen, 1988). The rhythm of slow-wave sleep is a mirror reflection of the activity rhythm and SWDs: low level during the dark phase and a sharp peak at the beginning of the light phase (Sei et al., 1997). This may be the reason why, if analyzed in 1 h resolution, high negative correlations were observed. Similar high correlations between SWDs on the one side and deep slow-wave sleep and active wakefulness (positive) were also seen in a 12 h sleep deprivation study during the light period in WAG/Rij rats (Drinkenburg et al., 1995). Data, in 2 h bins, showed that during sleep deprivation bins with the highest amount of wakefulness and, with the lowest amount of deep slow-wave sleep, were at the same times the bins with the highest number of SWDs. During the recovery period, characterized by a rebound of deep slow-wave sleep, bins with the highest amount of this type of sleep were bins with the lowest number of SWDs (Drinkenburg et al., 1995). The intimate co-occurrence between active wakefulness and SWDs as found in our present study might be due to the polyphasic character of the rat sleep and the short time lag between a period of active wakefulness, the transition to passive wakefulness and a drowsy state. The latter is the state at which SWDs are most prevalent. It is therefore also expected that changes in cortical excitability associated with the rhythm of active wakefulness might shape the rhythm of SWDs and be responsible for a close relationship between these two. Although the description of the temporal relationship of SWDs and sleep-wake states in 1 h bins is suited for the study of slow processes, it is quite long when considering the duration of SWDs or the duration of the sleep-wake cycle of the rat (Coenen and van Luijtelaar, 2003; Trachsel et al., 1991; van Luijtelaar and Bikbaev, 2007). In 6 min resolution, all states, except REM sleep, showed the highest cross-correlation coefficients at positive time lag 1, which means that the total time of a particular state in this bin might serve as a significant predictor of the time of SWDs in the following 6 min 78 epoch. Both active and passive wakefulness states, were positively cross-correlated with SWDs suggesting that their high amount will be followed by a relatively long total duration of SWDs, while negative cross-correlations between SWD with light and deep slow-wave sleep suggests the opposite. Interestingly, there was no sharp, dominant peak in cross-correlation coefficients with a resolution of 0.5 min, instead many cross-correlations were outside the confidence limits, indicating that the temporal relationship between SWDs on the one side and active wakefulness, light and deep slow wave sleep on the other is determined by their occurrence in 40 to 60 30-s epochs. Similarly as for 6 min resolution, both wakefulness states were crosscorrelated positively, both slow-wave sleep states negatively. Interestingly, small significant cross-correlation coefficients were additionally found for negative, more distant time lags suggesting an influence of SWDs themselves on the amount of sleep-wake states. Indeed, a shorter sleep cycle has been found in symptomatic WAG/Rij rats compared to age matched non-epileptic controls at the end of the light period suggesting a circadian vulnerability for mainly slow-wave sleep to be interrupted, in addition, the epileptic strain had shorter REM sleep episodes (Gandolfo et al., 1990; van Luijtelaar and Bikbaev, 2007). Clinical and experimental data yield also evidence for a reciprocal relationship between sleep and epilepsy. Not only may different sleep states alert the occurrence of seizures, but also seizures themselves disrupt the organization of the sleep-wake cycle (Baretto et al., 2002; Bazil et al., 2000; Maganti et al., 2005; Yi et al., 2012). The third main finding of the present study was a different strength of correlation and the temporal relationship between SWDs and vigilance states in the dark and light part of the 24 h day. Pearson’s correlation coefficients were higher in the light than in the dark phase for wakefulness (active and passive) and deep slow-wave sleep, irrespective of the resolutions. Also autocorrelation coefficients of 79 SWDs, passive wakefulness and slow-wave sleep states were higher or sometimes significant only in the light period. There were also noticeable differences between the light and dark period in spectral density, for some states the peaks were higher in the light than in the dark phase (passive wakefulness, light slow-wave sleep, deep slowwave sleep), for others it was reversed (SWDs, active wakefulness and REM sleep). Although there are clear differences in the percentage of time of sleep-wake states and in SWDs between light and dark, the amount of a certain sleep related variable does not determine the amplitude of the spectral density functions. Therefore, there must be different or additional mechanisms responsible for the differences in temporal organization between light and dark phase. In rats entrained to LD 12:12, 24 h rhythmicity in sleep-wake states is strong. However, in the absence of circadian influences, it is dominated by ultradian periodicities (Eastman et al., 1984; Ibuka et al., 1977). Stephenson et al. (2012) revealed that the period of an ultradian rhythm in wake, slow-wave and REM sleep in rats kept in 12:12 photoperiod varies randomly across a day within a range of 3 to 6 h. A phase-related difference was noticed: there was a more prominent rhythm during the light than during the dark phase. This was explained by the authors by a progressive desynchronization of ultradian oscillations throughout the circadian cycle (Stephenson et al., 2012). Our autocorrelation data also suggest that the occurrence of SWDs and almost all sleep-wake states have a stronger temporal control and that these temporal relationships are more stable during the light period compared to the dark period. Interestingly, higher spectral density functions that implies stronger rhythms were found in the dark period. The concept of the light onset entraining circadian timing system, that is gradually desynchronizing during the latter hours of the light phase and the subsequent dark period and giving more and more room across the evolving time for ultradian rhythms of sleep80 wake states to develop, might also serve as an explanation for the results obtained in the dark period: weaker and bidirectional temporal relationships between SWDs and sleep-wake states and more room for larger independent ultradian rhythms. It can be hypothesized that, if the presence of light at the circadian time 0 (CT 0) entrains two (or more) rhythms with different intrinsic period length each day to 24 h, then the dark phase would be the period during which the differences in temporal relationship between them will be most pronounced. The present study has some limitations. A major one is the arbitrary choice of time frames for analysis of temporal relationships. The number and strength of correlations and cross-correlations might depend on the bin size. However, preliminary analysis of the results included also 0.5 h and 10 min bins. The same tendencies were observed: an increase in data resolution was accompanied by a decrease of a value of coefficients. Another limitation might be the length of an epoch, based on which sleep-wake states were scored by the automated system. The program used 2 s epochs, different length e.g. 15 s might yield different results. On the other hand, the length of the smallest bin size in a final analysis was 30 s. The goal of the study was to investigate the temporal relationship between absence seizures and states of vigilance. We demonstrated that the temporal relation of SWDs with the different sleep-wake states depend on the bin size chosen. 1 h resolution of the data, typical for circadian research, gives opposite results as 30-s resolution but this regards only active wakefulness. The relationship between SWDs and deep slow-wave sleep is always negative, irrespective of the resolution. This suggests that deep slow wave sleep is a reliable factor determining the (non-)occurrence of absence seizures. The present study revealed also clear phase related differences in temporal relationship between variables investigated and their different organization in time and frequency domain. The light 81 phase of 12:12 photoperiod appeared to be the time during which stronger temporal relationship between SWDs and sleep-wake states took place. It is also the phase of higher stability of SWDs and vigilance states over time. The dark phase, however, is a period during which the relationship between absence seizures and sleep-wake states was weaker and bidirectional. The occurrence of SWDs and active wakefulness in that phase was dominated by many strong rhythmic oscillations in a range of short periodicities, while the spectral density of sleep-related states such as passive wakefulness, light and deep slow-wave sleep was lower. The results of their study suggest different mechanisms influencing the temporal organization of sleep, wakefulness and absence seizures in the light and the dark period. However, to reveal these mechanisms further investigations are needed. 5. Acknowledgements The authors would like to thank Heidi Huysmans and Dirk Nuyts for their excellent technical assistance and express the gratitude to Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium for supplying excellent facilities to perform the study. 6. Declaration of Interest The authors report no known conflict of interest. 7. References Ahnaou, A., Drinkenburg, W.H., 2011. Neuromedin U(2) receptor signaling mediates alteration of sleep-wake architecture in rats. Neuropeptides. 45, 165-174. 82 Badawy, R.A., Macdonell, R.A., Jackson, G.D., Berkovic, S.F., 2009. Why do seizures in generalized epilepsy often occur in the morning? Neurology. 73, 218-222. Barreto, J.R., Fernandes, R.M., Sakamoto, A.C., 2002. Correlation of sleep macrostructure parameters and idiopathic epilepsies. Arq Neuropsiquiatr. 60, 353-357. Bazil, C.W., Castro, L.H., Walczak, T.S., 2000. Reduction of rapid eye movement sleep by diurnal and nocturnal seizures in temporal lobe epilepsy. Arch. Neurol. 57, 363-368. Borbély, A.A., 1975. Circadian rhythm of vigilance in rat: Modulation by short light-dark cycles. Neurosci. Lett. 1, 67-71. Borbély, A.A., 1982. A two process model of sleep regulation. I Physiological basis and outline. Human Neurobiol. 1, 195-204. Coenen, A.M.L., van Luijtelaar, E.L.J.M., 2003. Genetic animal model for absence epilepsy: a review of the WAG/Rij strain of rats. Behav. Genet. 33, 635-55. Daan, S., Beersma, D.G., Borbély, A.A., 1984. Timing of human sleep: recovery process gated by a circadian pacemaker. Am. J. Physiol. 246, 161-183. Depaulis, A., van Luijtelaar, G., 2006. Genetic models of absence epilepsy in the rat, in: Pitkänen, A., Schwartzkroin, P.A., Moshe, S.L., (Eds.), Models of Seizures and Epilepsy. Elsevier Academic Press, San Diego, pp. 233-248. Drinkenburg, W.H., Coenen, A.M., Vossen, J.M., van Luijtelaar, E.L., 1991. Spike-wave discharges and sleep-wake states in rats with absence epilepsy. Epilepsy Res. 9, 218-224. Drinkenburg, W.H., Coenen, A.M., Vossen, J.M., van Luijtelaar, E.L., 1995. Sleep deprivation and spike-wave discharges in epileptic rats. Sleep 18, 252-256. Duncan, J.S., 1997. Idiopathic generalized epilepsy with typical absences. J Neurol. 244, 403-411. Eastman, C.I., Mistlberger, R.E., Rechtschaffen, A., 1984. Suprachiasmatic nuclei lesions eliminate circadian temperature and sleep rhythms in the rat. Physiol. Behav. 32, 357–368. Friedman, L., Bergmann, B.M., Rechtschaffen, A., 1979. Effects of sleep deprivation on sleepiness, sleep intensity and subsequent sleep in the rat. Sleep. 1, 369-391. Gandolfo, G., Romettino, S., Gottesmann, C., van Luijtelaar, G., Coenen, A.. 1990. Genetically epileptic rats show a pronounced intermediate stage of sleep. Physiol. Behav. 47, 213-215. 83 Halasz, P., Terzano, M.G., Parrino, L., 2002. Spike-wave discharge and the microstructure of sleep-wake continuum in idiopathic generalized epilepsy. Neurophysiol. Clin. 32, 38-53. Ibuka, N., Inouye, S.I., Kawamura, H., 1977. Analysis of sleep-wakefulness rhythms in male rats after suprachiasmatic nucleus lesions and ocular enucleation. Brain Res. 122, 33-47. Ibuka, N., Kawamura, H., 1975. Loss of circadian rhythm in sleep-wakefulness cycle in the rat by suprachiasmatic nucleus lesions. Brain Res. 96, 76-81. Karpova, A.V., Bikbaev, A.F., Coenen, A.M., van Luijtelaar, G., 2005. Morphometric Golgi study of cortical locations in WAG/Rij rats: the cortical focus theory. Neurosci. Res. 51, 119-128. Lannes, B., Micheletti, G., Vergnes, M., Marescaux, C., Depaulis, A., Warter, J.M., 1988. Relationship between spike-wave discharges and vigilance levels in rats with spontaneous petit mallike epilepsy. Neurosci. Letters. 94, 187-191. Lüttjohann, A., Zhang, S., de Peijper, R., van Luijtelaar, G., 2011. Electrical stimulation of the epileptic focus in absence epileptic WAG/Rij rats: assessment of local and network excitability. Neuroscience. 188, 125-134. Maganti, R., Sheth, R.D., Hermann, B.P., Weber, S., Gidal, B.E., Fine, J., 2005. Sleep architecture in children with idiopathic generalized epilepsy. Epilepsia. 46, 104109. Matzen, J., Buchheim, K., Holtkamp, M., 2012. Circadian dentate gyrus excitability in a rat model of temporal lobe epilepsy. Exp. Neurol. 234, 105-111. Meeren, H., van Luijtelaar, G., Lopes da Silva, F., Coenen, A., 2005. Evolving concepts on the pathophysiology of absence seizures: the cortical focus theory. Arch. Neurol. 62, 371-376. Midzyanovskaya I., Strelkov, V., van Rijn, C., Budziszewska, B., van Luijtelaar, E., Kuznetsova, G.,2006. Measuring clusters of spontaneous spike-wave discharges in absence epileptic rats. J. Neurosci. Methods. 154, 183-189. Ovchinnikov, A., Lüttjohann, A., Hramov, A., van Luijtelaar, G., 2010. An algorithm for realtime detection of spike-wave discharges in rodents. J. Neurosci. Methods. 194, 172-178. Paxinos, G., Watson, C., 1998. The rat brain in stereotaxic coordinates. Academic Press, San Diego. Polack, P.O., Guillemain, I., Hu, E., Deransart, C., Depaulis, A., Charpier, S., 2007. Deep layer somatosensory cortical neurons initiate spike-and-wave discharges in a genetic model of absence seizures. J. Neurosci. 27, 6590-6599. 84 Sei, H., Furuno, N., Morita, Y., 1997. Diurnal changes of blood pressure, heart rate and body temperature during sleep in the rat. J. Sleep Res. 6, 113-119. Simasko, S.M., Mukherjee, S., 2009. Novel analysis of sleep patterns in rats separates periods of vigilance cycling from long long durationwake events. Behav. Brain Res. 196, 228-236. Sitnikova, E., van Luijtelaar, G., 2007. Electroencephalographic characterization of spikewave discharges in cortex and thalamus in WAG/Rij rats. Epilepsia. 48, 2296-2311. Smyk, M.K., Coenen, A.M., Lewandowski, M.H., van Luijtelaar, G., 2011. Endogenous rhythm of absence epilepsy: relationship with general motor activity and sleepwake states. Epilepsy Res. 93, 120-127. Smyk, M.K., Coenen, A.M., Lewandowski, M.H., van Luijtelaar, G., 2012. Internal desynchronization facilitates seizures. Epilepsia. 53, 1511-1518. Stephan, F.K., Zucker, I., 1972. Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proc. Natl. Acad. Sci. USA. 69, 1583-1586. Stephenson, R., Lim, J., Famina, S., Caron, A.M., Dowse, H.B., 2012. Sleep-wake behavior in the rat: Ultradian rhythms in a light-dark cycle and continuous bright light. J. Biol. Rhythms. 27, 490-501. Suntsova, N., Kumar, S., Guzman-Marin, R., Alam, M.N., Szymusiak, R., McGinty, D., 2009. A role for the preoptic sleep-promoting system in absence epilepsy. Neurobiol. Dis. 36, 126-141. van Luijtelaar, E.L., Coenen, A.M., 1986. Two types of electrocortical paroxysms in an inbred strain of rats. Neurosci. Lett. 70, 393-397. van Luijtelaar, E.L.J.M., Coenen, A.M.L., 1988. Circadian rhythmicity in absence epilepsy in rats. Epilepsy Res. 2, 331-336. van Luijtelaar, E.L., van der Werf, S.J., Vossen, J.M., Coenen, A.M., 1991. Arousal, performance and absence seizures in rats. Electroecephalogr. Clin. Neurophysiol. 79, 430-434. van Luijtelaar, G., Coenen, A., 1993. The behavioural pharmacology of sleep, in van Haaren, F. (Eds.), Methods in behavioural pharmacology. Elsevier Science Publishers B.V., Amsterdam, pp 575-602. van Luijtelaar, E.L., Drinkenburg, W.H., van Rijn, C.M., Coenen, A.M., 2002. Rat models of genetic absence epilepsy: what do EEG spike-wave discharges tell us about drug effects? Methods Find. Exp. Clin. Pharmacol. 24, 65-70. 85 van Luijtelaar, G., Bikbaev, A., 2007. Midfrequency cortico-thalamic oscillations and the sleep cycle: genetic, time of day and age effects. Epilepsy Res. 73, 259265. van Twyver, H., 1969. Sleep patterns of five rodent species. Physiol. Behav. 4, 901905. Westmijse, I., Ossenblok, P., Gunning, B., van Luijtelaar, G., 2009. Onset and propagation of spike and slow wave discharges in human absence epilepsy: A MEG study. Epilepsia. 50, 2538-2548. Yi, P.L., Chen, Y.J., Lin, C.T., Chang, F.C., 2012. Occurrence of epilepsy at different zeitgeber times alters sleep homeostasis differently in rats. Sleep. 35, 16511665. 86 CHAPTER 3 Endogenous rhythm of absence epilepsy: relationship with general motor activity and sleep-wake states Published as Smyk MK, Coenen A, Lewandowski MH, van Luijtelaar G. 2011. Epilepsy Research. 2011. 93: 120-127. Abstract The rhythms of spontaneously occurring seizures (spike-wave discharges, SWD) and motor activity, as well as the relationship between SWD and sleep–wake states were investigated in the WAG/Rij rat model of absence epilepsy. In order to establish whether SWD are controlled by external (Zeitgebers) or by endogenous factors such as circadian influences or the state of vigilance, the study was performed in entrained and constant dim light conditions. EEG and motor activity were recorded in the 12:12 light–dark cycle and in constant dim light conditions. Circadian rhythmicity was found both for motor activity and the occurrence of SWD in conditions of entrainment. In constant dim light conditions also circadian rhythms emerged, however, the change in circadian parameters was opposite for the rhythm of SWD and motor activity. SWD were preceded mostly by passive wakefulness and by slow-wave sleep in both experimental conditions. It can be concluded that the rhythm of SWD seems to be generated and controlled by an endogenous mechanism distinct from that which controls the rhythm of motor activity. The relationship between SWD and sleep–wake states preceding their occurrences appeared to be unchanged, suggesting that the mechanism of generation of SWD is independent of the circadian timing system. Keywords: absence seizures; spike-wave discharges; WAG/Rij rats; circadian rhythmicity; motor activity; sleep–wake states 1. Introduction The majority of mammalian physiological and behavioural processes are controlled by a circadian timing system. The light-dark 89 cycle, the most prominent synchronizer of biological rhythms, entrains a circadian master clock, the hypothalamic suprachiasmatic nuclei (SCN), to a 24-h day (Reppert and Weaver, 2001). The SCN control other clocks through multiple neuronal connections and humoral factors, and these clocks express overt circadian rhythms (Buijs and Kalsbeek, 2001; Morin and Allen, 2006). These various oscillators or rhythms show a phase-relationship under entrained conditions. However, the SCN generates a rhythm with a period length different from 24 h in absence of time cues (Bünning, 1964; Herzog and Schwarts, 2002). Not only the period may change, but also parameters such as amplitude and mean as well as the relationships between rhythms and even uncoupling of distinct circadian rhythms may occur. This phenomenon is called “internal desynchronization” and has been reported in humans and animals (Weaver, 1979; Aschoff and Weaver, 1981; Erkert, 2000; Aguzzi et al., 2006). Internal desynchronization is thought to deliver evidence supporting a “multiple oscillatory theory” of the mammalian circadian timing system. Also pathological processes such as the timing of epileptic seizures show daily rhythmic fluctuations. Investigations initiated by Gowers (1885), who classified seizures as diurnal, nocturnal and diffuse, led to the conclusion that distinct circadian patterns of the occurrence of epileptic discharges are determined by the type of epilepsy syndrome, both in humans and in animal models (Gowers, 1885; Langdon-Down and Brain, 1929; Pavlova et al., 2004; Durazzo et al., 2008; Hofstra et al., 2009; Quigg et al., 1998). It seems that the sleep-wake or rest-activity cycle plays a major role among the different circadian factors that ultimately influence the timing of seizures (Halasz, 1991). Changes in neuronal excitability associated with these rhythms determine the susceptibility for seizures to occur at specific or preferred states of vigilance. 90 Idiopathic generalized epilepsy (IGE) with typical absences belongs to a group of epilepsy syndromes that is strongly affected by the sleep-wake and rest-activity cycle. Indeed a 24-h rhythm has been identified for the occurrence of absence seizures (Kellaway et al., 1980). Absences are characterized by the presence of bilateral, synchronous, generalized spike-wave discharges (SWD) in the EEG against a normal background activity. The frequency of the discharges is about 3 Hz and differs slightly among particular syndromes (Panayiotopoulos, 1994; Duncan, 1997). Slow-wave sleep, quiet wakefulness (e.g. physical and mental inactivity associated with drowsiness), light slow-wave sleep and transitions, mainly awakenings from slow-wave sleep are the most favourable states for SWD to occur, both in humans and in animal models (Coenen et al., 1991; Drinkenburg et al., 1991; van Luijtelaar and Bikbaev, 2007). SWD hardly occur during the highly desynchronized EEG activity such as active wakefulness and REM sleep (Dinner, 2002; Halasz et al., 2002). Rats of the WAG/Rij strain are a well known, validated, genetic animal model of absence epilepsy sharing common behvioural, electroencephalographic and pharmacological characteristics with the human disorder (Coenen and van Luijtelaar, 2003; Depaulis and van Luijtelaar, 2006). The distribution of the 7 – 11 Hz SWD show a clear circadian variation (van Luijtelaar and Coenen, 1988; van Luijtelaar et al., 2001), with a maximum during the early hours of the dark phase and a minimum at the beginning of the light phase, when rats have the highest amount of deep slow-wave sleep. The latter state is not favourable for the occurrence of SWD (Drinkenburg et al., 1991). Therefore, it is likely that the distribution of SWD is a direct consequence of the distribution of the vigilance states and the presence of Zeitgebers. The purpose of the present study was to verify the putative endogenous character of the rhythm of SWD. Considering that the 91 probability of the occurrence of SWD is higher during states of physical inactivity, emphasizing a role of vigilance in the generation of absences, the relationship between sleep-wake states including motor activity and the occurrence of SWD was additionally investigated in order to verify whether the absence of Zeitgeber light on and off interferes with the dependency observed under conditions of entrainment. 2. Material and methods 2.1. Animals Seven adult (eleven months old), male WAG/Rij rats, born and raised in the laboratory of the Donders Centre for Cognition of the Radboud University Nijmegen, served as subjects. Animals were maintained on a 12:12 light-dark cycle (lights on at 6:00 am) with food and water available ad libitum. All experimental procedures were approved by the Animal Ethical Committee of the Radboud University Nijmegen. 2.2. Surgery Rats were implanted with a standard three-channel electrode set (MS 333/1-A, Plastic One Inc., Roanoke, VA, USA) under isoflurane anaesthesia. Two active electrodes were put on the surface of primary motor and visual cortex. Coordinates of the electrodes (with skull surface flat and bregma zero-zero) were: AP +2.0, ML 3.5 and AP -6.0, ML 4.0, respectively. The reference electrode was put over the cerebellum. Animals were allowed to recovery for 14 days after the surgery. 92 2.3. EEG and activity recordings Each animal was individually placed in sound-attenuated, ventilated recording cage and connected with a computer-based data acquisition system (Dataq Instruments, Akron, OH, USA). Rats were connected to a swivel which allowed them to move freely in their cages. The EEG signals were amplified, band pass filtered between 1 and 100 Hz, and sampled at 200 Hz. General motor activity was measured by a passive infrared sensor (PIR) mounted 40 cm above the bottom of the animal’s cage and connected to the data acquisition system. This system, which has been used earlier and validated against visual observation, measures the total body movements over a period of time, showing a large amplitude during intense body movements such as digging, rearing, grooming and exploration, and an almost zero amplitude during behavioural immobility. Animals were allowed to habituate to the recording cages for at least 2 h before the recordings started. 2.4. Experimental design Animals were kept on the 12:12 light-dark cycle (light intensity: 60 lux) which constitutes the condition of entrainment. An overview of the design of the experiment is presented in Fig. 1. The light regime was changed after 10 days into constant dim light (5-6 lux), on which rats were maintained for the following 20 days. EEG recordings were made continuously during the last four days of the entrained condition and during the last seven days of the constant dim light condition (from day 23 to day 30). Motor activity recordings were made throughout the whole experiment (Fig. 1). Food and water were always available ad libitum, the presentation of fresh water and the addition of new rat chow in the constant dim light condition was done at irregular time 93 points, just as the cleaning of cages. This was done only once a week to minimize intervention and prevent synchronization. Figure 1. Experimental design 2.5. Data analysis Preliminary PIR recordings were visually inspected to define an activity threshold value. The number of episodes above the threshold (= active behavior) was counted automatically from the PIR recordings. Recordings were divided into 6 min bins. Each bin with a predominance of (> 50%) above the threshold was scored as 1. The value 0 was given to bins with the predominance of passive behavior. Activity data were divided into 10 day segments: the first of which represents 10 days spent in the entrained condition, the second representing the last 10 days of 20 days spent in constant dim light conditions. The period, mean and amplitude of the activity rhythms of both segments were analysed by means of the Cosinor method, which yields also a measure of the robustness of the rhythm (the percentage of variance accounted for by the rhythm). 94 In order to compare sleep, motor activity, SWD in the active and passive phases of the entrained and constant dim light periods, the beginning and the end of the active period in the entrained and constant dim light conditions was established. The onset of darkness was taken as the beginning of the active phase for the entrained condition. The onset of the activity rhythm in the constant dim light condition was based on the outcomes of the Cosinor method which gives the accurate estimation of the period length. In addition, visual inspection of the activity rhythm over days showed marked and long lasting activity bouts after a period of behavioral inactivity. The beginning of the activity bouts over days was used to estimate the beginning of the active period. Next the circadian day was split in two equal parts (the active and passive phase). It was noticed that the beginning of the passive period always coincided with long lasting period of behavioral inactivity. EEGs were recorded during 4 days in the entrained condition and during 7 days of the constant dim light condition. SWD were identified off-line according to well-known criteria (van Luijtelaar and Coenen, 1986). The Cosinor analysis was applied on 4 and 7 day segments of the data collected in the entrained and constant dim light conditions, respectively (data resolution: 6 min, number of SWD per 6 min block), to estimate the circadian period, mean, amplitude and robustness of the SWD rhythm. Sleep-wake states were identified off-line by visual inspection of the EEG and PIR recordings from the last day of the entrained and the last day of constant dim light condition, commonly used criteria were used (van Twyver, 1969; van Luijtelaar and Coenen, 1993). The second and penultimate hour of the dark and the light phase of the 12;12 light-dark cycle and corresponding hours of the active and the passive phase of the constant dim light condition were analysed. Four sleep-wake states were scored: 95 active wakefulness, passive wakefulness, slow-wave sleep and REM sleep. To determine the preference of the occurrence of SWD for a particular state of vigilance, every 5 s preceding and following each SWD were analysed. Results were subsequently corrected with regards to the distribution of the sleep-wake states over the analysed hours, as proposed by Drinkenburg et al. (1991). 2.6. Statistical analysis Statistical analysis was performed in Statistica (StatSoft, Inc., USA). Multivariate analysis of variance was first used to estimate differences in parameters between light regimes, only in case of significant condition effect, Student’s t-tests for dependent samples were used as post hoc tests. Differences were considered as significant at p < 0.05. 3. Results In the entrained condition (24 h, 12:12 light-dark), a rhythm of motor activity was found with a period length of 24.03 ± 0.04 h. Animals were more active during the dark phase than during the light phase of the 12:12 light-dark cycle (p < 0.001). Multivariate analysis of variance showed significant differences between experimental condition for both motor activity and SWD rhythm (F = 84.84, df = 4.3, p < 0.01 and F = 10.91, df = 4.3, p < 0.05, respectively). Subsequent post hoc tests showed that the period of motor activity rhythm increased to 24.63 ± 0.08 h (p < 0.01), while the mean, amplitude and robustness of the rhythm decreased (all p’s < 0.05) in the constant dim light. The number of activity counts was higher for the active than for the passive phase of the constant dim light condition (p < 0.001). A comparison between the entrained and 96 the constant dim light condition revealed that animals were less active in the active phase and more active in the passive phase of the constant dim light condition (both p’s < 0.05). Data are presented in Table 1. Entrained Period [h] Meana Amplitude Robustnessb Constant dim light 24.03 ± 0.04 24.63 ± 0.08* 0.20 ± 0.03 0.16 ± 0.06* 0.18 ±0.01 0.11 ± 0.01* 8.58 ± 1.59 4.28 ± 0.64* 342.43 ± 46.40 241.86 ± 33.18* 85.86 ± 17.30 109.43 ± 17.37* Activity counts Dark/active Light/passive Table 1. Mean and SEM of the various parameters of the motor activity rhythm in the entrained and constant dim light condition. a An estimate of central tendency of the distribution of values of an oscillating variable. b The percentage of variance accounted for by the rhythm. * Entrained vs dim light condition, p < 0.05 The occurrence of SWD in the entrained phase showed also a clear, circadian rhythm. The minimum occurred during the first 2 h of the light phase. Then the number gradually increased to reach a maximum at the end of that phase. At the beginning of the dark phase a small drop occurred followed by an increase and then again a continuous decrease (Fig. 2). There were no differences in the number of SWD between the light and the dark phase of the 24-h rhythm, as well as between the active and the passive phase of the SWD rhythm recorded in the constant dim light condition. However, the number of SWD was higher in the active phase in comparison with the dark phase 97 of the entrained condition (p < 0.05). The period of the SWD rhythm in constant dim light condition was shorter than 24 h (22.93 ± 0.53), although the difference just failed to reach significance. However, each animal showed a significant (it means that a significant amount of variation across time points was explained by the fitted cosinus), circadian rhythm in the occurrence of SWD under the constant dim light condition. Significant changes were also found for the mean and amplitude of the rhythm, which were larger in the constant dim light condition (p < 0.05; p < 0.001, respectively), as well as for the robustness, which was smaller than in the entrained condition (p < 0.05). Results are presented in Table 2. Entrained Period [h] Meana 24.01 ± 0.04 Constant dim light 22.93 ± 0.53 1.41 ± 0.18 1.82 ± 0.15* 5.45 ±0.48 8.32 ± 0.68* 4.44 ± 0.70 1.57 ± 0.29* Dark/active 631.43 ± 81.08 919.28 ± 65.89* Light/passive 738.43 ± 77.38 865.71 ± 90.38* Amplitude Robustnessb SWD number Table 2. Mean and SEM of the various parameters of the SWD rhythm in the entrained and constant dim light condition. a An estimate of central tendency of the distribution of values of an oscillating variable. b The percentage of variance accounted for by the rhythm. * Entrained vs dim light condition, p < 0.05 98 Figure 2. Double plotted graph of 4 days of SWD (mean number and SEM of SWD per hour) activity (one X-axis represents two days [48 h], the activity of the second day is plotted again as the first day in the next X-axis). The dark phase of the 12:12 light-dark cycle is marked in grey 99 The comparison of parameters of the activity and SWD rhythm in the constant dim light condition revealed significant differences. The period length of the SWD rhythm was shorter and the rhythm less robust (p’s < 0.05), while the amplitude and the mean of the SWD rhythm was higher in comparison to the activity rhythm (both p’s < 0.001). The distribution of particular states of vigilance in the entrained condition is presented in Table 3. Active wakefulness, dominating at the beginning of the dark phase, decreased gradually till the beginning of the light phase, in which it reached its lowest level. At the end of the light phase the amount of active wakefulness slightly increased. The same pattern was also observed for passive wakefulness, while the course of the slow-wave sleep was opposite. The lowest amount of slow-wave sleep was observed at the beginning of the dark phase, subsequently followed by a gradual increase till the beginning of the light phase when it reaches its maximum. Contrary to active and passive wakefulness, the amount of slow-wave sleep decreased at the end of the light phase. The percentage of REM sleep gradually increased during the investigated hours. Active wakefulness in the constant dim light condition was found to be the only state of vigilance, which preserved its pattern observed in the entrained condition. The course of the remaining sleep-wake states was altered. The lowest amount of passive wakefulness was found at the end of the active phase, followed by an increase at the beginning of the passive phase and a subsequent decrease at the end. A minimal amount of slow-wave sleep was recorded, the same as in the entrained condition, at the beginning of the active phase. The maximum was shifted from the beginning of the passive phase to the end of the active phase, after which a gradual decrease was observed. A slight alteration was also found for REM sleep. The same pattern for three investigated periods (the entire active phase and the beginning 100 of the passive phase) was preserved, with the exception of the end of the passive phase in which a decrease was observed. The distribution of the particular states of vigilance recorded in the constant dim light condition is presented in Table 3. Dark phase Beginning Light phase End Beginning End Entrained condition Active wakefulness 60.22 ± 3.61 38.21 ± 9.22 12.13 ± 4.18 15.56 ± 3.84 Passive wakefulness 18.26 ± 1.96 13.08 ± 3.38 5.00 ± 1.24 10.45 ± 1.66 Slow-wave sleep 19.10 ± 3.64 40.70 ± 8.54 73.33 ± 5.37 61.47 ± 2.82 REM sleep 2.42 ± 1.22 8.01 ± 3.21 9.53 ± 1.75 12.52 ± 1.72 Constant dim light condition Active wakefulness 54.16 ± 7.41 26.54 ± 8.88 23.02 ± 6.16 39.56 ± 5.53 Passive wakefulness 12.41 ± 1.14 6.77 ± 1.63 15.61 ± 6.93 10.95 ± 2.60 Slow-wave sleep 30.26 ± 5.92 59.65 ± 7.23 49.69 ± 7.86 40.58 ± 6.33 REM sleep 3.17 ± 2.01 7.04 ± 2.10 11.68 ± 2.55 8.90 ± 1.30 Table 3. Mean and SEM of the percentage of particular states of vigilance during investigated hours in the entrained and constant dim light conditions A comparison made between the entrained and the constant dim light condition revealed that in the active phase of the constant dim light condition the percentage of passive wakefulness was lower (p < 0.05). In the passive phase of the constant dim light condition the 101 percentage of active wakefulness was higher (p < 0.05), whereas the percentage of slow-wave sleep was lower (p < 0.05). Passive wakefulness and slow-wave sleep have been found to dominate among states of vigilance preceding the occurrence of SWD in the entrained condition. 39% and 35% of SWD recorded were preceded by passive wakefulness and by slow-wave sleep, respectively, active wakefulness (15%) and REM sleep (11%) were the least probable states. This tendency was preserved in the constant dim light condition. 47% of SWD were preceded by passive wakefulness, 32% by slow-wave sleep, 17% by active wakefulness and 4% by REM sleep. No significant differences between the entrained and the constant dim light condition were found. Data are presented in Fig. 3. Passive wakefulness in the entrained condition followed the occurrence of SWD most frequently (71%), active wakefulness, with a probability of 17%, was the second state. Slow-wave sleep and REM sleep were recorded after SWD in 9% and 3% of cases, respectively. The same relationship was found in the constant dim light condition for two sleep-wake states: passive wakefulness and REM sleep. Passive wakefulness was the most and REM sleep the least probable to occur after SWD (62% and 9%, respectively). Opposite to the entrained condition, slow-wave sleep was found to be the second most frequent state following the occurrence of SWD (18%), active wakefulness was the third (11%). Data are presented in Fig. 3. Statistical analysis revealed that the probability that SWD was followed by slow-wave sleep is significantly higher in the constant dim light condition than in the entrained condition (p < 0.05). 102 Figure 3. States of vigilance preceding (A and C) and following (B and D) the occurrence of SWD in the entrained (A and B) and constant dim light conditions (C and D). AW = active wakefulness, PW = passive wakefulness, SWS = slowwave sleep, REM = REM sleep. SWS after SWD occurred more often in the constant dim light condition, * p < 0.05 4. Discussion In the present study the rhythm of absence seizures, the rhythm of general motor activity and the relationship between absence seizures and states of vigilance in WAG/Rij rats were investigated under two different experimental conditions: the condition of entrainment to the 12:12 light-dark cycle, and the condition of constant dim light. Here it is shown for the first time that there is an endogenous rhythm of absence seizures, given that the circadian rhythmicity in the occurrence of SWD is still present in the absence of the most important Zeitgeber. Moreover, under the constant dim light 103 condition significant changes in various parameter of the rhythm of SWD and in general motor activity are observed, while additionally internal desynchronization between these rhythms is observed. Contrary to these changes, the well established relationship between the occurrence of SWD and states of vigilance in not altered by the constant environment. Regardless of environmental conditions, SWD are most frequently occurring during passive wakefulness and slowwave sleep. The present results confirm previous observations of circadian rhythmicity in rats’ motor activity (Büttner and Wollnik, 1984; Ray et al., 2004; Refinetti, 2006). Animals are more active during the dark than during the light phase in the entrained condition. The constant dim light alters parameters of the motor activity rhythm. The period of the rhythm becomes longer than 24 h, in agreement with “Aschoff’s rule” stating that, in nocturnal animals maintained on constant light, an increase in light intensity corresponds with an increase in the period length of the rhythm. It can be concluded that a slight, but significant, increase in the period length observed in the present experiment is caused by the presence of constant low intensity light on which animals are maintained (Aschoff, 1960; Daan and Pittendrigh, 1976; Fukuhara et al., 2005). The remaining parameters such as the mean, amplitude and robustness of the rhythm are found to be decreased in comparison with the condition of entrainment. An inhibitory effect of light on the activity of nocturnal animals is well established (Redlin, 2001). A longlasting, continuous exposure to dim light has been reported to reduce the amplitude and the mean of activity rhythm with a possible disruption of the circadian pattern (Borbély and Neuhaus, 1978; Albers et al., 1981; Eastman and Rechtschaffen, 1983; Ikeda and Inoue, 1998). A circadian rhythmicity in the occurrence of SWD in WAG/Rij rats is found in the entrained condition. The observed pattern shares 104 common characteristics with previous reports (van Luijtelaar and Coenen, 1988). The circadian distribution is a direct consequence of the changes in sleep-wake states over the course of the 24-h day (Drinkenburg et al., 1991). Light slow-wave sleep, passive wakefulness and transitions between different states of vigilance have been shown to promote the occurrence of SWD, while deep slow-wave sleep, REM sleep and active wakefulness are states exerting opposite effects (Lannes et al., 1988; Coenen et al., 1991; Drinkenburg et al., 1991). In both studies the nadir of the rhythm was found to be located during the first hours of the light phase, which correlates with a high amount of deep slow-wave sleep at that time of the light-dark cycle (Coenen et al., 1991; Drinkenburg et al., 1991). A slight difference was noted for the maximum of the rhythm: it occurred at the end of the light phase, while van Luijtelaar and Coenen (1986) pointed out that this maximum emerged at the early hours of the dark phase. Despite the high number of SWD in the dark phase, a decline was observed just at the beginning of it. For nocturnal animals kept in laboratory conditions, turning off the light is a strong signal to become behaviorally active. Therefore, the characteristic peak of activity at the beginning of the dark period could be responsible for such a decline, as active wakefulness suppresses the occurrence of SWD (Coenen et al., 1991; Drinkenburg et al., 1991). The changes in the SWD rhythm that took place in the constant dim light condition show an opposite direction in comparison with changes in the rhythm of motor activity. The period length of the absence epilepsy rhythm becomes shorter than 24 h. Although this change is not significant according to parametric tests for the group as unity, each animal showed a clear circadian rhythm with a new, shorter period length in the range of 20.6 – 23.9 h. A non-parametric sign test confirmed that the period length was indeed shorter in constant dim light. A circadian rhythm of limbic seizures was first described by Quigg 105 et al. (2000) in a model for temporal lobe epilepsy: the seizures occurred in a phasic nonrandom pattern both under entrainment and under constant darkness with a period length of about 24 h. Similar to the present results, the number of seizures, as well as the amplitude of the rhythm increased in an environment lacking external time cues. Contrary to a decrease in the amplitude and in the mean of the motor activity rhythm, the corresponding parameters of the rhythm of SWD increased. Motor activity and waking are suppressed in the condition of continuous light (Borbély and Neuhaus, 1978). This suppression can be a putative cause for the increase in the number of SWD considering the intimate negative relationship between vigilance and arousal, and the occurrence of SWD. Moreover, it has been shown that the condition of constant light disrupts the sleep-wake pattern. A significant increase of slow-wave and REM sleep is found under constant light, together with an elevated number of REM sleep episodes (Borbély and Neuhaus, 1978). Similar results were obtained by Ikeda et al. (2000), additionally pointing towards a redistribution of the amount of particular sleep stages in the active and the passive phase. In the present study, dim light alters the course, as well as the amount of sleep-wake states. The divergence of the period length under constant dim light between motor activity, with a significant increase, and the SWD rhythm, with a tendency to decrease, suggests distinct oscillators responsible for their regulation. Others have found dissociation between circadian rhythms in continuous dim light in monkeys and rats (Erket, 2000; Aguzzi et al., 2006). Studies devoted to the process of desynchronization of oscillators are considered as important evidence in multiple oscillatory theory of mammalian circadian timing system. These oscillators may reside in or outside the SCN (Pittendrigh and Daan, 1976; Shinohara et al., 1995; Jagota et al., 2000). 106 A common finding for both the motor activity and the SWD rhythm, is a gradually decreasing robustness after the transfer to the constant dim light condition. Both rhythms show the same tendency, however, the motor activity rhythm was found to be significantly more robust throughout the whole experiment than the SWD rhythm. This suggest that the motor activity rhythm is governed by a stronger oscillator. Differences in robustness of the rhythms of wake and body temperature under long-term continuous illumination were earlier reported: the wake rhythm, thought to be driven by a weaker oscillator, lost its organization earlier than the body temperature rhythm (Eastman and Rechtschaffen, 1983). Although the 22.9 h rhythm of SWD remained identifiable, it was also noticed that it became less organized. The motor rhythm kept its organization better. Both rhythms showed a diminished robustness, reflecting a decrease in the strength of the circadian timing system. Therefore, it seems that under entrained conditions, the structure of the SWD rhythm is influenced by changes in sleep-wake states and in the amount and timing of activity over 24-h day (Coenen et al., 1991; Drinkenburg et al., 1991). A disrupted organization of general motor activity, expressed by a decreased robustness of its rhythm, together with a distinct oscillator acting independently from the oscillator controlling the timing of SWD, might be responsible for the gradual change of the structure of the SWD rhythm under the constant dim light condition. In the present study, the majority of SWD was preceded by passive wakefulness and slow-wave sleep, while active wakefulness and REM sleep were rarely recorded just before SWD, consistent with previous findings from both animal and human studies (Lannes et al., 1988; Drinkenburg et al., 1991; Halasz et al., 2002). The relationship between SWD and states of vigilance is the same in both: the entrained and constant dim light condition, strongly suggesting that the 107 dependency of generation of SWD on particular state of vigilance is not influenced by circadian factors. SWD are often followed by active and passive wakefulness, this awaking effect of SWD observed in the present experiment supports earlier findings (Drinkenburg et al., 1991). Here it is found that the probability that SWD are followed by slow-wave sleep is higher in constant dim light than in entrained condition, while the sleep-wake state preceding the onset of SWD is not changed. This implies that the awaking properties of SWD are diminished in the constant dim light condition. The sleep-wake cycle of these epileptic rats has been found to be disrupted: besides shorter and many aborted REM sleep periods (Suntsova et al., 2009; Gandolfo et al., 1991), the sleep cycle is shortened. However, the sleep cycle disturbances are significant only at a specific time of day, pointing towards a circadian factor in sleep disturbances (van Luijtelaar and Bikbaev, 2007). It seems that the tendency of SWD to exert awaking effect is reduced by uncoupling the SWD and motor activity rhythms and by a decrease in the robustness of these rhythms. In conclusion, this study demonstrates that the timing of SWD in the entrained conditions is due to photic synchronizing stimuli and it is shaped by the motor activity rhythm and the momentary state of vigilance. It also confirmed a true circadian nature of the rhythm of absence seizures. Considering an opposite tendency in the characteristics of rhythms under the constant dim light condition, it can be concluded that the SWD rhythm might be driven by a circadian oscillator distinct form that of the general motor activity rhythm. The relationship between states of vigilance and the mechanisms of the generation of SWD is preserved in the constant dim light condition suggesting, that this mechanism is independent from circadian modulation. However, the awaking effect of SWD became weaker. The uncoupling of SWD and motor activity rhythms during the constant dim 108 light condition allows that the SWD rhythm loses some of its robustness. 5. Acknowledgements The study is supported by a fellowship from Radboud University Nijmegen. The authors would like to thank Gerard van Oijen, Pascal de Water for technical and Saskia Hermeling and Hans Krijnen for biotechnical assistance. 6. References Aguzzi J, Bulloc, N.M., Tosini, G. 2006. Spontaneuos internal desynchronization of locomotor activity and body temperature rhythms from plasma melatonin rhythm in rats exposed to constant dim light. J Circadian Rhythms 4, 4-6. Albers HE, Gerall AA, Axelson JF. 1981. Circadian rhythm dissociation in the rat: effect of long-term constant illumination. Neurosci Lett 25, 89-94. Aschoff J. 1960. Exogenous and endogenous components in circadian rhythms. Cold Spring Harbor. Symp. Quant. Biol. 25, 11-28. Aschoff J, Wever R. 1981. The circadian system of man. (Eds.), Handbook on Behavioural Neurobiology. Biological Rhythms. London: Plenum, 311-331. Borbély AA, Neuhaus HU. 1978. Circadian rhythm of sleep and motor activity in the rat during skeleton photoperiod, continuous darkness and continuous light. J Comp Physiol 128, 37-46. Buijs RM, Kalsbeek A. 2001. Hypothalamic integration of central and peripheral clocks. Nat Rev Neurosci 2, 521-6. Bünning E. 1964. The Physiological Clock. Springer Verlag, Berlin. Büttner D, Wollnik F. 1984. Strain-differentiated circadian and ultradian rhythms in locomotor activity of the laboratory rat. Behav Genet 14, 2. Coenen AM, Drinkenburg WH, Peeters BW, Vossen JM, van Luijtlaar EL. 1991. Absence epilepsy and the level of vigilance in rats of the WAG/Rij strain. Neurosci Biobehav Rev 15, 259-63. Coenen AML, van Luijtelaar ELJM. 2003. Genetic animal model for absence epilepsy: a review of tha WAG/Rij strain of rats. Behav Genet 33, 635-55. 109 Daan S, Pittendrigh CS. 1976. A functional analysis of circadian pacemakers in nocturnal rodents. III Heavy water and constant light: homeostasis of frequency? J Comp Physiol 106, 267-290. Depaulis A, van Luijtelaar ELJM. 2006. Genetic models of absence epilepsy in the rat. In A. Pitkänen, P.A. Schwartzkroin, & S.L. Moshe (Eds.), Models of Seizures and Epilepsy, 233-248. Amsterdam: Elsevier Academic Press. Dinner DS. 2002. Effect of sleep on epilepsy. J Clin Neurophysiol 19, 504-13. Drinkenburg WH, Coenen AM, Vossen JM, van Luijtelaar EL. 1991. Spike-wave discharges and sleep-wake states in rats with absence epilepsy. Epilepsy Res. 9, 218-24. Duncan JS. 1997. Idiopathic generalized epilepsieswith typical absences. J Neurol 244, 403-411. Durazzo TS, Spencer SS, Duckrow RB, Novotny EJ, Spencer DD, Zaveri HP. 2008. Temporal distribution of seizure occurrence from various epileptogenic regions. Neurology 70, 1265-1271. Eastman C, Rechtschaffen A. 1983. Circadian temperature and wake rhythmsof rats exposed to prolonged continuous illumination. Physiol Behav 31, 417-427. Erket HG. 2000. Internal desynchronization of the circadian activity and feeding rhythm in an owl monkey (Aotus lemurinus griseimembra): a case study. Chronobiol Int 17, 147-53. Fukuhara C, Aguzzi J, Bullock N, Tosini G. 2005. Effect of long-term exposure to constant dim light on the circadian system of rats. Neurosignals 14, 117-125. Gowers W. 1885. Course of epilepsy. (Eds.), Epilepsy and Other Chronic Convulsive Diseases: Their Causes, Symptoms and Treatment. New York: William Wood, 157–164. Halasz P. 1991. Sleep, arousal and electroclinical manifestations of generalized epilepsy with spike wave pattern. Epilepsy Res. Suppl. 2, 43-8. Halasz P, Terzano MG, Parrino L. 2002. Spike-wave discharge and the microstructure of sleep-wake continuum in idiopathic generalized epilepsy. Neurophysiol Clin 32, 38-53. Herzog ED, Schwartz WJ. 2002. A neuronal clockwork for encoding circadian time. J Appl Physiol 92, 401-8. Hofstra WA, Spetgens WPJ, Leijten FSS, van Rijen PC, Gosselaar P, van der Palen J, de Weerd AW. 2009. Diurnal rhythms in seizures detected by intracranial electrocorticographic monitoring: An observational study. Epilepsy Behav 14, 617-621. 110 Ikeda M, Inoue S. 1998. Simultaneous recording of circadian rhythms of brain and intraperitoneal temperatures and locomotor and drinking activities in the rat. Biol. Rhythm. Res. 29, 142-150. Ikeda M, Sagara M, Inoue S. 2000. Continuous exposure to dim illumination uncouples temporal pattern of sleep, body temperature, locomotion and drinking behaviour in the rat. Neurosi. Lett 279, 185-189. Jagota A, de la Iglesia HO, Schwartz WJ. 2000. Morning and evening circadian oscillations in the suprachiasmatic nucleus in vitro. Nat Neurosci 3, 372-376. Kellaway P, Frost Jr JD, Crawley JW. 1980. Time modulation of spike-and-wave activity in generalized epilepsy. Ann Neurol 8, 491-500. Langdon-Down M. Brain W. 1929. Time of day in relation to convulsions in epilepsy. Lancet 1029-1032. Lannes B, Micheletti G, Vergnes M, Marescaux Ch, Depaulis A, Warter JM. 1988. Relationship between spike-wave discharges and vigilance levels in rats with spontaneous petit mal-like epilepsy. Neurosci. Letters 94, 187-191. Morin LP, Allen CN. 2006. The circadian visual system. Brain Res Rev 51, 1-60. Panayiotopoulos CP. 1994. The clinical spectrum of typical absence seizures and absence epilepsies. (Eds.), Idiopathic generalized epilepsies: clinical, experimental and genetic aspects. Malafosse, A., Genton, P., Hirsch, E., Marescaux, C., Broglin, D., Bernasconi, R. John Libbey & Company Ltd. 75-85. Pavlova MK, Shea SA, Bromfield EB. 2004. Day/night patterns of focal seizures. Epilepsy Behav 5, 44-49. Pittendrigh CS, Daan S. 1976. A functional analysis of circadian pacemakers in nocturnal rodents. V. Pacemaker structure: a clock for all seasons. J. comp. Physiol. 106, 333-355. Quigg M, Straume M, Menaker M, Bertram EH III. 1998. Temporal distribution of partial seizures: comparison of an animal model with human partial epilepsy. Ann Neurol 43, 748-755. Quigg M, Clayburn H, Straume M, Menaker M, Bertram III EH. 2000. Effects of circadian regulation and rest-activity state on spontaneous seizures in rat model of limbic epilepsy. Epilepsia 41, 502-509. Ray B, Mallick HN, Kumar VM. 2004. Changes in thermal preference, sleep-wakefulness, body temperature and locomotor activity of rats during continuous recording for 24 hours. Behav Brain Res. 154, 519-26. Redlin U. 2001. Neural basis and biological function of masking by light in mammals: suppression of melatonin and locomotor activity. Chronobiol Int 18, 737-758. 111 Refinetti R. 2006. Variability of diurnality in laboratory rodents. J Comp Physiol A 192, 701-714. Reppert SM, Weaver DR. 2001. Molecular analysis of mammalian circadian rhythms. Annu Rev Physiol 63, 647-76. Shinohara K, Honma S, Katsuno Y, Abe H, Honma K. 1995. Two distinct oscillators in the rat suprachiasmatic nucleus in vitro. Proc. Natl. Acad. Sci. 92, 7396-7400. Suntsova N, Kumar S, Guzman-Marin R, Alam MN, Szymusiak R, McGinty D. 2009. A role for the preoptic sleep-promoting system in absence epilepsy. Neurobiol Dis 36, 126-141. van Luijtelaar EL, Coenen AM. 1986. Two types of electrocortical paroxysms in an inbred strain of rats. Neurosci Lett. 70, 393-397. van Luijtelaar ELJM, Coenen AML. 1988. Circadian rhythmicity in absence epilepsy in rats. Epilepsy Res. 2, 331-336. van Luijtelaar G, Coenen A. 1993. The behavioural pharmacology of sleep. In van Haaren F. (Ed.), Methods in behavioural pharmacology, 575-602, Amsterdam: Elsevier Science Publishers B.V. van Luijtelaar G, Budziszewska B, Jaworska-Feil L, Ellis J, Coenen A, Lasoń W. 2001. The ovarian hormones and absence epilepsy: a long-term EEG study and pharmacological effects in a genetic absence epilepsy model. Epilepsy Res. 46, 225-39. van Luijtelaar G, Bikbaev A. 2007. Midfrequency cortico-thalamic oscillations and the sleep cycle: genetic, time of day and age effects. Epilepsy Res. 73, 259-265. van Twyver H. 1969. Sleep patterns of five rodent species. Physiol Behav 4, 901-905. Wever RA. 1979. The circadian system of man. Springer Verlag 276, New York. 112 CHAPTER 4 Internal desynchronization facilitates seizures Published as Smyk MK, Coenen A, Lewandowski MH, van Luijtelaar G. Epilepsia. 2012. 53: 1511-8. Abstract Purpose: The occurrence of spike-wave discharges (SWDs) in WAG/Rij rats is modulated by the circadian timing system and is shaped by the presence of a light–dark cycle, motor activity, and state of vigilance. Here it is investigated whether the response to a phase shift is different between the SWDs and general motor activity rhythm. The process of re-entrainment of both rhythms and its effect on number of absences was compared after a phase shift in the light–dark cycle, a condition known to induce internal desynchronization in the circadian timing system. Methods: Chronic electroencephalographic and motor activity recordings were made in adult WAG/Rij rats, kept in the 12:12 h light– dark cycle. After four baseline days, rats were exposed to an 8-h phase delay by shifting the light onset. Recordings were continuously made for another 10 consecutive days. Key Findings: An immediate effect of the phase shift on both rhythms was observed: the acrophases were 7.5 h advanced. Next, they gradually returned to the baseline level, however, with a different speed. The more robust motor activity rhythm stabilizes first, whereas the weaker rhythm of SWDs adapted more slowly. The phase shift caused a prolonged aggravation of epileptic activity, observed mostly during the light phase. Significance: Different speed and character of re-entrainment suggests that the occurrence of seizures and motor activity are controlled by distinct circadian oscillators. The prolonged increase in absences after the phase shift has immediate practical consequences. Keywords: absence epilepsy; WAG/Rij rats; genetic model; spike-wave discharges; phase shift; circadian rhythm 115 1. Introduction The rhythmic expression of physiologic functions is the result of complex interactions between the external and internal environment and the circadian timing system. A direct connection with the retina allows the master circadian pacemaker, the suprachiasmatic nucleus of the hypothalamus (SCN), to respond to cyclic changes in environmental illumination, one of the most potent synchronizers of biological rhythms (Morin & Allen, 2006). The SCN entrained by a lightdark cycle and supported by other time cues, governs dependent clocks so that a stable phase relationship between various oscillators is reached (Buijs & Kalsbeek, 2001). However, sudden phase shifts in the light-dark cycle are able to disturb this synchrony. The speed of entrainment to a new photoperiod is different for various oscillators; it is the ventrolateral, photosensitive part of the SCN that synchronizes first and other follow (Yamazaki et al., 2000; Nagano et al., 2003; Lee et al., 2009). Internal desynchronization during the process of reentrainment is also thought to be direct cause of jet lag: a state of fatigue, poor performance, and sleep disturbances reported by travelers, who rapidly cross several time zones (Haimov & Arendt, 1999; Waterhouse et al., 2007). Circadian control over physiologic and behavioral processes in mammals is well-documented (Bünning, 1964). In addition, some pathologic phenomena such as epileptic seizures tend to occur daily, in a nonrandom fashion (Gowers, 1885; Langdon-Down & Brain, 1929; Quigg et al., 1998). The distribution of seizures across a 24-h day depends on the type of epileptic syndrome, and can be also influenced by other circadian variables such as the sleep-wake or rest-activity cycle, both in humans and animal models (Kellaway et al., 1980; van Luijtelaar & Coenen, 1988; Coenen et al., 1991; Drinkenburg et al., 116 1991; Halasz, 1991; Halasz et al., 2002; Pavlova et al, 2004; Durazzo et al., 2008; Hofstra et al., 2009). Absence seizures are manifested by the presence of bilateral, synchronous, generalized spike-wave discharges (SWDs) in the electroencephalogram (EEG), accompanied by behavioral arrest, reduced consciousness, and decreased responsiveness (Depaulis & van Luijtelaar, 2006). The distribution of SWDs across 24-h period in WAG/Rij rats, a well-known and validated genetic model of absence epilepsy (Coenen & van Luijtelaar, 2003; van Luijtelaar & Sitnikova, 2006), is rhythmically organized, with a maximum at the beginning of the dark phase and a minimum localized during the first 2 hours of the light phase (van Luijtelaar & Coenen, 1988; Drinkenburg et al., 1995; Smyk et al., 2011). It is assumed that at least three main factors contribute to this structure: photic entrainment, momentary state of vigilance, and motor activity. Previous findings have demonstrated that the SWD rhythm is generated endogenously, since it is still present in rats maintained in a condition of constant dim light, in which the circadian timing system lacks time cues. In such a condition, the number of seizures was elevated, however, the relationship between the occurrence of SWDs and the state of vigilance was not affected. In addition, desynchronization between the rhythms of SWDs and general motor activity was observed, which suggests the existence of distinct oscillators for both rhythms (Smyk et al., 2011). The present study aimed to investigate whether two oscillators with different circadian properties also differ in the entrainment to an 8-h phase shift. More specifically, circadian characteristics of SWDs including number and duration and motor activity rhythm, and their speed of entrainment were determined before, during and 10 days after a sudden phase shift, which was induced by extending the dark period. 117 2. Methods 2.1. Animals Eight adult WAG/Rij rats (age 18 months, weight 336 ± 37 g [mean + SEM]), born and raised in the laboratory of the Department of Biological Psychology, Donders Centre for Cognition, Radboud University Nijmegen, were used in the experiment. Animals were housed in pairs in standard polycarbonate cages and maintained in a 12:12 h light-dark cycle (lights on at 8 a.m.) with food and water available ad libitum. All experimental procedures were approved by the Radboud University Nijmegen Animal Ethical Committee. 2.2. Surgery Stereotactic surgery was performed under general isoflurane anesthesia. Each rat was implanted with a standard three-channel electrode set (MS 333/1-A; Plastic One Inc., Roanoke, VA, U.S.A.). Two active electrodes were placed on the surface of primary motor and visual cortex (coordinates with the skull flat and bregma zero-zero: AP +2.5, ML 3.5 and AP – 6.0, ML 4.0, respectively). The reference electrode was placed over the cerebellum. Rats were housed individually during 14 days of a recovery period after the surgery, as well as throughout the entire experiment. 2.3. EEG and motor activity recordings Rats were placed in sound-attenuated, individually ventilated recording cages and connected with a computer-based data acquisition system (Dataq Instruments, Akron, OH, U.S.A.), by means of cables and swivels, which allowed animals to move freely around their cages. 118 The EEG signals (differential recordings) were amplified, band pass filtered between 1 – 100 Hz, and sampled at 256 Hz. General motor activity was recorded by a passive infrared sensor (PIR), mounted 40 cm above the bottom of cages and connected to the data acquisition system. A signal recorded showed high amplitude in response to large body movements, such as locomotion, food intake, or grooming and almost zero amplitude during behavioral immobility. Animals had been habituated to cages and cables 24 h before the experiment started. 2.4. Experimental design Baseline EEG and general motor activity recordings were made for 4 days in the 12:12 h light-dark cycle, with lights on at 8 a.m. and lights off at 8 p.m. White light with the intensity of 60 lux at the bottom of animals cage was provided during the light phase, whereas the dark phase constituted of total darkness. The dark phase of the fifth day was extended from 12 to 20 h (lights off at 8 p.m., lights on at 4 p.m., instead of 8 a.m., next day), in order to produce a light phase delay. During and after the phase delay, recordings were continuously made for 10 days in the 12:12 h light-dark cycle, with lights on at 4 p.m. and lights off at 4 a.m. (postshift days). 2.5. Data analysis The analyses were identical as in our previous experiment (Smyk et al., 2011). In brief, PIR recordings were divided into 6 min. bins. The mean of an absolute value of the raw signal was calculated for each bin and expressed subsequently as a percentage of the mean of all recorded days. A cosine wave with a fixed period length of 24 h was fitted to each baseline and postshift day (each day = 240 data points, one point = values for one 6-min bin), to estimate circadian 119 parameters of the rhythm of general motor activity: the mean, amplitude, acrophase, and robustness (the percentage of the variance accounted for by the rhythm) (program: Cosinor, available at http:/www.circadian.org/softwar.html), next to whether a significant amount of variation over time was obtained. Values obtained for four baseline days were subsequently averaged. SWDs were identified offline according to well-known criteria (van Luijtelaar & Coenen, 1986; Ovchinnikov et al., 2010). The total number and mean duration of SWDs were calculated for each experimental day. Circadian parameters of the rhythm of SWDs were estimated independently from the rhythm of general motor activity, however, the procedure was the same. A cosine wave with a fixed period length of 24 h was fitted to each baseline and post-shift day (each day = 240 data points, one point = the number of SWDs for one 6-min bin). The number, mean duration and circadian parameters obtained for the baseline days were averaged. The rate of the entrainment of the rhythms of general motor activity and SWDs was determined by inspection of individual acrophase data, and expressed as the number of days after which the acrophase was the closest to the baseline level. In addition, the extended dark period (8 h) preceding the light phase shift was analyzed. Each hour of the extended darkness was compared with corresponding hours of the beginning of the light and the dark phase from the baseline. Parameters compared were the number of SWDs and the level of activity. 2.6. Statistical analysis Statistical analysis was performed in SPSS (18th edition; SPSS Inc., Chicago, IL, U.S.A.). Repeated measures analyses of variance (ANOVA) were used including orthogonal trend analysis to estimate day 120 effects in circadian parameters, in SWD number and mean duration, and the level of activity between baseline and postshift days and between the light and dark period. The same repeated measures was used to estimate differences in the number of SWDs and level of activity in the extended dark period, the beginning of the light phase and the beginning of the dark phase. Least significant difference (LSD) and Student t-tests were used as post hoc tests to delineate differences between days and to estimate differences between light and dark periods and in the rate of the entrainment of the rhythms of general motor activity and SWDs. Differences were considered to be significant at p-level < 0.05. 3. Results 3.1. General motor activity A clear, 24-h circadian rhythmicity was found in general motor activity in the baseline condition. Animals were more active during the dark than during the light phase of the 12:12 h light-dark cycle (149.6 ± 8.8 vs. 67.3 ± 3.4 %, respectively, representing the mean level of activity of all experimental days (MLA) of the dark and light periods, p < 0.001). The two-way repeated measures ANOVA for the total activity score in the dark and light period over all recorded days (baseline and 10 days after the phase shift) showed a significant day (F = 3.21, d.f. 10,70, p < 0.01), a light-dark (F = 249.89 d.f. 1,7, p < 0.0001) and an interaction effect (F = 23.43, d.f. 10,70, p < 0.001). Post hoc tests showed that the phase-related level of activity of the baseline was reversed on the first postshift day, since the rats were now more active during the light phase (100.6 ± 6.2 % vs. 88.2 ± 8.3 % of the MLA, p < 0.05). The commonly found activity difference between light and 121 dark was restored on the third postshift day and remained present from that moment onward (all p < 0.05 or < 0.001). Comparisons between days showed that the level of nocturnal activity was reduced for 8 days (days 1 - 6, 8: all p < 0.001, day 7: p < 0.05), whereas the level of diurnal activity was elevated for 4 days (day 1, 2, 4: all p < 0.001, day 3: p < 0.05). Circadian parameters of the rhythm of general motor activity were also changed over time (F = 60.66, d.f. 10, 70, p < 0.001). The acrophase of the rhythm was advanced immediately after the phase shift (from 259.9 ± 8.7 to 157.0 ± 10.8o, p < 0.01). The return to the baseline level took 6 days (days 1 - 5: all p < 0.01, day 6: p < 0.05). Both the mean and the amplitude of the rhythm changed over time (F = 2.93, d.f. 10, 70, p < 0.01 and F = 2.51, d.f. 10, 70, p < 0.05, respectively). The mean decreased from the first till the eight postshift day (day 1-3, 5: all p < 0.01, day 6 and 8: all p < 0.05) with an exception of the fourth and seventh day; on these days the effects were in the same direction but failed to reach significance. The amplitude was decreased on the first, second and fourth postshift day (58.4 ± 6.6 vs. 43.1 ± 7.1, 46.0 ± 5.7 and 46.7 ± 9.5, respectively, day 1: p < 0.01, day 2, 4: all p < 0.05). A slight decrease over days was observed for the robustness of the rhythm (F = 2.21, d.f. 10, 70, p < 0.05); the post hoc tests showed a lower robustness on the first and fourth post-shift day (all p < 0.05). General motor activity data are presented in Fig. 1. 122 Figure 1. Mean and SEM of the level of general motor activity in the light and dark phase (A), the mean (B) and the acrophase (C) of the rhythm of general motor activity. Y axis: (A and B) percentage of the mean level of activity of all days recorded (MLA), degrees (360 degrees = 24 h, 0 – 180 degrees – the light phase, 181 – 360 degrees – the dark phase); X axis: experimental days divided into baseline period (4 days) and postshift period (10 days), day on 123 which the phase shift was produced is not included. * p < 0.05, LSD post hoc test after ANOVA, mean baseline level versus postshift day 3.2. Spike-wave discharges A 24-h rhythm in the occurrence of SWDs was found in the baseline condition. The minimum of the rhythm was localized at the beginning of the light phase, whereas the acrophase occurred during early hours of the dark phase. A mean number of SWDs 463.2 ± 113.2, lasting on average 6.6 ± 1.1 s, was recorded. SWDs were more numerous during the dark than the light phase of the 12:12 h lightdark cycle (296.2 ± 92.9 vs. 186.6 ± 53.9, p < 0.01). The two-way repeated measures ANOVA on number of SWDs of all recording days showed a day effect that could be described by a quadratic trend (Fquad = 8.72, d.f. 1,7, p < 0.01) and a day x time of day interaction (F = 5.09, d.f .10,70, p < 0.001). Subsequent one-way ANOVAs for the light and dark period separately, showed a day effect (F = 4.92, d.f. 10, 70, p < 0.001). The number of seizures was elevated after the phase shift, starting from the first till the seventh postshift day (days 1, 4, 6: all p < 0.05, day 2, 3, 5, 7: all p < 0.01). The oneway ANOVA on the number of SWDs in the dark period showed a day effect (F = 2.27, d.f. 10, 70, p < 0.05), which could be further described by the presence of a linear rising trend (Flin = 10.38, d.f. 1, 7, p < 0.05). The number of SWDs on the first postshift day in the dark phase tended to be decreased, but failed to reach significance (242.6 ± 96.8 vs. 296.2 ± 92.9). The SWD data clearly demonstrate that the overall increase in the number of seizures occurred in the light phase. The phase shift induced also a phase reversal between the light and dark period in the number of SWDs. Differences in SWDs between the light and dark period, present during the baseline day (t = 2.66, d.f. 7, p < 0.05) were only restored on the eighth, (t = 2.39, p < 0.05), 124 ninth (t = 3.39, p < 0.01) and tenth (t = 1.97, p < 0.10, all d.f. = 7) postshift days. The acrophase of the rhythm of SWDs was significantly affected by the phase shift. An immediate advance was observed on the first postshift day (221.8 ± 31.6 vs. 108.6 ± 30.2o, p < 0.001). A significant time effect was found (F=8.34, d.f. 10, 70, p<.001). The return to the baseline level took 3 days (day 2 and 3, all p < 0.001), however, a significant difference was also observed on the seventh postshift day (p < 0.001). SWDs data are presented in Figure 2. The other circadian parameters were not significantly changed. Orthogonal trend analyses showed a significant quadratic trend for the mean (F = 8.89, d.f. 1, 7, p < 0.05) and amplitude (F = 8.60, d.f. 1, 7, p < 0.05) of the SWD rhythm, suggesting that the initial increase was followed by a decrease. Subsequent post hoc tests failed to find clear differences between days. This might be due to the fact that in 4 of 8 animals the procedure of fitting the 24-h cosine line to the data set from some of the postshift days, mostly from the early period after the phase shift, failed to reach significance. This implies that the temporal organization of the SWD rhythm, as found during the baseline period, was less obvious after the phase delay. The mean duration of SWDs was also affected by the phase shift: a significant day effect was found (F = 3.13, d.f. 10,70, p < 0.01); post hoc tests showed that the SWDs lasted longer on postshift days 1, 2 and 4 and 10 compared to base-line (all p < 0.05). 125 Figure 2. Mean and SEM of the number of SWDs in the light and dark phase (A), the mean duration of SWDs (B), the acrophase of the SWD rhythm (C) and the rate of entrainment of the rhythms of general motor activity and SWDs (D). Y axis: (A) number of SWDs, (B) seconds, (C) degrees (360 degrees = 1h, 0 – 180 degrees – the light phase, 181 – 360 degrees – the dark phase), (D) day; X axis: (A,B and C) experimental days divided into baseline period (4 days) and postshift period (10 days), day on which the phase shift was produced is not included. * p < 0.05, LSD post hoc tests after ANOVA, mean baseline level versus postshift day (A, B and C), * p < 0.05, Student’s t-test (D) 126 3.2.1. Spike-wave discharges The same ANOVA on the number of SWDs showed a significant period effect (F = 13.74, d.f. = 2, 14, p < 0.001). Post hoc tests showed that there were no significant differences between the extended dark and the light phase of the baseline condition; however, the rats had less (all p < 0.05) SWDs during the extended dark phase and the light phase of the baseline condition compared to the dark phase of the baseline condition. 4. Discussion The process of re-entrainment to an 8-h light phase delay was investigated in a genetic animal model of absence epilepsy. Circadian parameters of the rhythm of SWDs and the rhythm of general motor activity, as well as the number of SWDs and the level of activity, were compared before, during and after the time shift. The most prominent changes were an advance of the acrophase of both rhythms, followed by a gradual synchronization to the new photoperiod, as well as an aggravation of epileptic activity. The speed and character of entrainment of the investigated rhythms were found to be different. General motor activity entrained faster and steadily, while the rhythm of SWDs, temporarily synchronized after the third postshift day, completed the process significantly later. The effect of an abrupt shift of the light-dark cycle on the circadian rhythm of motor activity is well established. The magnitude of changes caused by the shift depends on its amount and direction (Wever, 1966). Generally, disturbances caused by phase delays are managed faster than those caused by phase advances. The process of reentrainment after a 6-, 8- or 10-h phase delay took 2, 4 or 6 days, respectively, in albino strains of rats (Takamure et al. 1991, Marumoto 127 et al, 1996; Yamazaki et al., 2000; Nagano et al., 2003). In the present study, the acrophase of general motor activity of the rats reached the baseline level on the sixth postshift day, which, although 2 days longer and perhaps due to the complex phenotype (epilepsy and comorbidity of depression), remains in accordance with the above-cited studies. Interestingly, there were no differences in the speed of entrainment or any other circadian parameters between rats with the highest and lowest number of absences. Apart from the timing of the acrophase, also the phase-related distribution of motor activity was changed. For 8 days after the phase shift, nocturnal activity of WAG/Rij rats was decreased, whereas the level of daytime activity was elevated for 4 days. Similar results were obtained in mice after a light-dark cycle reversal: nocturnal activity was decreased 4 - 8 days after the shift, whereas a significant difference between nocturnal and diurnal activity was reestablished on days 4 - 7 dependent on the strain (Kopp et al. 1998). In addition, Wistar rats subjected to a 10-h phase delay showed an increased daytime and reduced nocturnal activity. The phase-related distribution of activity reached the baseline level on sixth day after the shift (Nagano 2003). Despite small differences, the process of synchronization of the general motor activity to the shifted photoperiod in WAG/Rij rats shares characteristics in common with other strains of nocturnal rodents. The entrainment of the circadian rhythm of absence seizures to a phase shift in the light-dark cycle was not previously investigated, neither in man nor in rats. During the baseline condition, a clear 24-h rhythm of SWDs was observed in all WAG/Rij rats. The shape of the rhythm (localization of minimum and maximum, phase related number of SWDs), together with its circadian parameters, confirmed previous findings (van Luijtelaar & Coenen, 1988; Smyk et al., 2011). A response to an 8-h phase delay shared some common features with 128 the process of entrainment of the general motor activity rhythm. In both rhythms, an advance of the acrophase and a redistribution of the phase-related motor and epileptic activity took place. However, the speed and the nature of the entrainment of the motor and SWD rhythm were rather different. The acrophase of the rhythm of SWD was immediately advanced on the first postshift day and was localized in the middle of the light phase, instead of the beginning of the dark phase. The entrainment to the new photoperiod took 8 days, during which a transient synchronization on the fourth postshift day was observed, followed by a subsequent shift from the baseline level, and finally, by a completion of the process. Therefore, the rate of the entrainment and the course of synchronization were different between the rhythms of SWDs and general motor activity. In our previous study performed in constant condition, an internal desynchronization between these two rhythms was observed, suggesting distinct mechanisms responsible for their entrainment (Smyk et al., 2011). The phenomenon of desynchronization between circadian oscillators and rhythms after the phase shift has also been described by others. The process of reentrainment of central and peripheral oscillators in transgenic rats was completed at first in the SCN, and then in peripheral tissues (Yamazaki et al., 2000). The SCN of the rat, known to consist of the photosensitive ventrolateral and nonphotosensitive dorsomedial part, followed the transient internal desynchronization as a result of the phase shift (Nagano et al., 2003; Lee et al., 2009). The ventrolateral SCN shifted immediately, whereas the dorsomedial SCN required 6 days to reach the new phase. Differences in the rate of entrainment within these two parts of the SCN are reflected in desynchronization between the rhythms of slow-wave sleep and REM sleep controlled by these morphologically and functionally distinct parts. The proper phase angle between these two rhythms of sleep stages is restored on 3 - 5 129 days after the shift (Lee et al., 2009). Different rate of entrainment has been reported also for the rhythms of core body temperature and locomotor activity in mice subjected to an 8-h phase advance (Satoh et al., 2006). The results obtained in the present study support the hypothesis about independent slave oscillators for the rhythms of SWDs and motor activity (Smyk et al., 2011). Both rhythms are controlled by and synchronized to the master circadian pacemaker under the 12:12 h light-dark cycle that controls the rhythms, whereas they act independently to reestablish a proper phase relationship with the SCN and the Zeitgebers after the phase shift. It was previously observed that these two rhythms/oscillators differ in their robustness, both, in the entrained and in the constant condition (Smyk et al., 2011). The rhythm of general motor activity was found to be more robust than the SWD rhythm. These differences might contribute to the different course of the entrainment to the phase shift observed in the present study. It is proposed that a strong, robust rhythm of motor activity synchronized faster and steadily, whereas the weaker rhythm of SWDs entrained slower. Moreover, the phase shift itself was found to disrupt the rhythmic occurrence of seizures as some animals lost their rhythmicity in SWDs during the early stages of entrainment, similarly as this was reported for peripheral oscillators (Yamazaki et al., 2000). In the present study, in contrast to the outcomes of the analysis of SWDs, a non significant circadian pattern for motor activity was never observed at any of the stages of entrainment. After 8 hr light phase delay, epileptic activity, measured by the number and mean duration of SWDs, was found to be elevated. The prolonged increase in the number of seizures was accompanied by a redistribution of the phase-related number of SWDs, a phenomenon described above for the rhythm of general motor activity. However, contrary to motor activity, the phase inversion lasted longer, whereas 130 the significant difference between phases was reestablished later. The steady increase in SWDs was also found when rats were kept in constant dim light conditions compared to the baseline 12:12 h lightdark cycle (Smyk et al. 2011). This increase was partially attributed to a declined motor activity, since SWDs are most numerous during immobile behavior (Drinkenburg et al., 1991). In the present study, the increase of the number of SWD was seen mainly in the light phase. At the same time, the diurnal level of motor activity was above baseline level, which suggests that other factors contribute to this phenomenon, since high levels of locomotor activity are incompatible with the occurrence of absence seizures. Melatonin and corticosterone are endogenous substances known to affect SWDs (Sandyk, 1992; Kldiashvili et al., 2001; Schridde & van Luijtelaar, 2004). Both conditions: constant dim light and phase shift, are known to alter circadian rhythms and the level of their secretion. The melatonin rhythm in the rat is disrupted or diminished in constant dim light and also during a few days after the phase shift (Humlova & Illnerova, 1990; Fukuhara et al., 2005; Liu & Borjigin, 2005; Aguzzi et al., 2006). In addition, the level of corticosterone in the rat is elevated and the circadian rhythm of its secretion is changed after an experimentally induced let lag (Mohawk et al., 2007). Melatonin and corticosterone have been reported to exert opposite effect on absence seizures in WAG/Rij rats: attenuating and aggravating, respectively. However, the role of these hormones in the process of entrainment of the SWD rhythm to the new photoperiod is not known and should be clarified in future studies. The analysis of eight additional hours of darkness preceding the phase shift, revealed no differences in the level of activity and the number of SWDs between this period and the corresponding hours of the light phase. Regardless of the photic information, rats showed passive behavior. The phase shift day consisted of 32 hours, 12 light 131 and 20 dark hours, which is far outside the range of entrainment for the rat (Stephan, 1983; Madrid et al., 1998). When the limit is exceeded, the circadian timing system cannot synchronize to the Zeitgeber and starts free running (Bünning, 1964; Herzog & Schwartz, 2002). Diurnal behavior in both motor and epileptic activity, during the extended dark period, is a sign of a free-running state of the circadian timing system of WAG/Rij rats. After an artificially long, 32-h day, the 12:12 h light-dark cycle has been regularly imposed on the rats till the end of the experiment. Because the light-dark cycle is one of the strongest Zeitgeber, rats gradually synchronize to the new photoperiod. In conclusion, the present study demonstrates that the circadian rhythms of SWDs and general motor activity are controlled independently by distinct slave oscillators. Rhythms are under the control of the master circadian pacemaker (SCN) and are coupled to each other in the entrained condition. The phase shift induces internal desynchronization and the rhythms show different rates of entrainment. The strong robust motor activity rhythm stabilizes first, while the weaker rhythm of SWD adapts more slowly. Internal desynchronization is thought to be responsible for complaints associated with jet lag. Here it is shown that absences increase especially during the day as a consequence to an experimental manipulation resembling jet lag. This finding might be important for people with epilepsy, undergoing rapid time shifts. 5. Acknowledgments The study is supported by a fellowship from Radboud University Nijmegen. The authors would like to thank Gerard van Oijen for technical and Saskia Menting-Hermeling and Hans Krijnen for biotechnical assistance. 132 6. Disclosure None of the authors has any conflict of interest to disclose. We confirm that we have read the Journal’s position on issues involved in ethical publication and affirm that this report is consistent with those guidelines. 7. References Aguzzi J, Bulloc NM, Tosini G (2006) Spontaneuos internal desynchronization of locomotor activity and body temperature rhythms from plasma melatonin rhythm in rats exposed to constant dim light. J Circadian Rhythms 4: 4-6. Buijs RM, Kalsbeek A (2001) Hypothalamic integration of central and peripheral clocks. Nat Rev Neurosci. 2: 521-526. Bünning E (1964) The Physiological Clock. Springer Verlag, Berlin. Coenen AM, Drinkenburg WH, Peeters BW, Vossen JM, van Luijtlaar EL (1991) Absence epilepsy and the level of vigilance in rats of the WAG/Rij strain. Neurosci. Biobehav. Rev. 15: 259-263. Coenen AML, van Luijtelaar ELJM (2003) Genetic animal model for absence epilepsy: a review of tha WAG/Rij strain of rats. Behav. Genet. 33: 635-655. Depaulis A, van Luijtelaar ELJM (2006) Genetic models of absence epilepsy in the rat. In Pitkänen A, Schwartzkroin PA, Moshe SL (eds) Models of Seizures and Epilepsy, Elsevier Academic Press, San Diego, pp. 233-248. Drinkenburg WH, Coenen AM, Vossen JM, van Luijtelaar EL (1991) Spike-wave discharges and sleep-wake states in rats with absence epilepsy. Epilepsy Res. 9: 218-224. Drinkenburg WH, Coenen AM, Vossen JM, van Luijtelaar EL (1995) Sleep deprivation and spike-wave discharges in epileptic rats. Sleep 18: 252-256. Durazzo TS, Spencer SS, Duckrow RB, Novotny EJ, Spencer DD, Zaveri HP (2008) Temporal distribution of seizure occurrence from various epileptogenic regions. Neurology 70: 1265-1271. Fukuhara C, Aguzzi J, Bullock N, Tosini G (2005) Effect of long-term exposure to constant dim light on the circadian system of rats. Neurosignals 14: 117-125. 133 Gowers W (1885) Course of epilepsy. In Gowers W (eds) Epilepsy and Other Chronic Convulsive Diseases: Their Causes, Symptoms and Treatment, William Wood, New York, pp.157–164. Haimov I, Arendt J (1999) The prevention and treatment of jet lag. Sleep Med Rev. 3: 229-240. Halasz P (1991) Sleep, arousal and electroclinical manifestations of generalized epilepsy with spike wave pattern. Epilepsy Res Suppl. 2: 43-48. Halasz P, Terzano MG, Parrino L (2002) Spike-wave discharge and the microstructure of sleep-wake continuum in idiopathic generalized epilepsy. Neurophysiol Clin. 32: 38-53. Herzog ED, Schwartz WJ (2002) A neuronal clockwork for encoding circadian time. J Appl Physiol. 92: 401-498. Hofstra WA, Spetgens WPJ, Leijten FSS, van Rijen PC, Gosselaar P, van der Palen J, de Weerd AW (2009) Diurnal rhythms in seizures detected by intracranial electrocorticographic monitoring: An observational study. Epilepsy Behav. 14: 617-621. Humlova M, Illnerova H (1990) Rate of re-entrainment of circadian rhythms to advances of light-dark cycles may depend on ways of shifting the cycles. Brain Res. 531: 304-306. Kellaway P, Frost Jr JD, Crawley JW (1980) Time modulation of spike-and-wave activity in generalized epilepsy. Ann Neurol. 8: 491-500. Kldiashvili S, Melechov M, Morenkov E, Coenen AML, van Luijtelaar G, Chepurnov SA (2001) Effects of the sleep-wake rhythm modulating hormone melatonin on absence seizures in the EEG of rats. In van Bemmel AL, Beersma DGM, Hofman WF, Ruigt GSF, Vos PJE (eds) Sleep-wake research in the Netherlands, Vol 12, Dutch Society for Sleep-Wake Research, Leiden, pp. 58-61. Kopp C, Vogel E, Rettori MC, Delagrange P, Guardiola-Lemaître B, Misslin R (1998) Effects of a daylight cycle reversal on locomotor activity in several inbred strains of mice. Physiol Behav. 63: 577-585. Langdon-Down M, Brain W (1929) Time of day in relation to convulsions in epilepsy. Lancet 1: 1029-1032. Lee ML, Swanson BE, de la Iglesia HO (2009) Circadian timing of REM sleep is coupled to an oscillator within the dorsomedial suprachiasmatic nucleus. Curr Biol. 19: 848-852. Liu T, Borjigin J (2005) Reentrainment of the circadian pacemaker through three distinct stages. J Biol Rhythms. 20: 441-450. 134 Madrid JA, Sánchez-Vázquez FJ, Lax P, Matas P, Cuenca EM, Zamora S (1998) Feeding behavior and entrainment limits in the circadian system of the rat. Am J Physiol. 275: 372-383. Marumoto N, Murakami N, Kuroda H, Murakami T (1996) Melatonin accelerates reentrainment of circadian locomotor activity rhythms to new light-dark cycles in the rat. Jpn J Physiol. 46: 347-51. Mohawk JA, Pargament JM, Lee TM (2007) Circadian dependence of corticosterone release to light exposure in the rat. Physiol Behav. 92: 800-806. Morin LP, Allen CN (2006) The circadian visual system. Brain Res Rev. 51: 1-60. Nagano M, Adachi A, Nakahama K, Nakamura T, Tamada M, Meyer-Bernstein E, Sehgal A, Shigeyoshi Y (2003) An abrupt shift in the day/night cycle causes desynchrony in the mammalian circadian centre. J Neurosci. 23: 6141-6151. Ovchinnikov A, Lüttjohann A, Hramov A, van Luijtelaar G (2010) An algorithm for realtime detection of spike-wave discharges in rodents. J Neurosci Methods. 194: 172-178. Pavlova MK, Shea SA, Bromfield EB (2004) Day/night patterns of focal seizures. Epilepsy Behav. 5: 44-49. Quigg M, Straume M, Menaker M, Bertram EH III (1998) Temporal distribution of partial seizures: comparison of an animal model with human partial epilepsy. Ann Neurol. 43: 748-755. Sandyk R (1992) Melatonin and petit-mal epilepsy: an hypothesis. Int J Neurosci. 65: 83-90. Satoh Y, Kawai H, Kudo N, Kawashima Y, Mitsumoto A (2006) Temperature rhythm reentrains faster than locomotor rhythm after a light phase shift. Physiol Behav. 88: 404-10. Schridde U, van Luijtelaar G (2004) Corticosterone increases spike-wave discharges in a dose- and time-dependent manner in WAG/Rij rats. Pharmacol Biochem Behav. 78: 369–375. Smyk MK, Coenen AM, Lewandowski MH, van Luijtelaar G (2011) Endogenous rhythm of absence epilepsy: relationship with general motor activity and sleep-wake states. Epilepsy Res. 93: 120-127. Stephan FK (1983) Circadian rhythms in the rat: constant darkness, entrainment to T cycles and to skeleton photoperiods. Physiol Behav. 30: 451-462. Takamure M, Murakami N, Takahashi K, Kuroda H, Etoh T (1991) Rapid reentrainment of the circadian clock itself, but not the measurable activity rhythms to a new light-dark cycle in the rat. Physiol Behav. 50: 443-449. 135 van Luijtelaar EL, Coenen AM (1986) Two types of electrocortical paroxysms in an inbred strain of rats. Neurosci Lett. 70: 393-397. van Luijtelaar ELJM, Coenen AML (1988) Circadian rhythmicity in absence epilepsy in rats. Epilepsy Res. 2: 331-336. van Luijtelaar G, Sitnikova E (2006) Global and focal aspects of absence epilepsy: the contribution of genetic models. Neurosci Biobehav Rev. 30: 983- 1003. Waterhouse J, Reilly T, Atkinson G, Edwards B (2007) Jet lag: trends and coping strategies. Lancet 369: 1117-1129. Wever R (1966) The duration of re-entrainment of circadian rhythms after phase shifts of the Zeitgeber. A theoretical investigation. J Theor Biol. 13: 187-201. Yamazaki S, Numano R, Abe M, Hida A, Takahashi R, Ueda M, Block GD, Sakaki Y, Menaker M, Tei H (2000) Resetting central and peripheral circadian oscillators in transgenic rats. Science 288: 682-685. 136 CHAPTER 5 Re-entrainment of sleep-wake states and absence seizures after a phase delay Abstract Rapid changes in photoperiod lead to desynchonization of circadian rhythms, its consequences require a considerable amount of time to overcome. Timed administration of melatonin or its agonists was found beneficial in accelerating re-synchronization. The aim of the present study was to investigate whether and how the various sleepwake states and absence seizures re-adapt to an 8h phase delay and whether the melatonin agonist agomelatine affects the speed of reentrainment of the different sleep-wake states. Two experiments were conducted: an acute pharmacological study towards the effects of various doses of agomelatine on sleepwake states and absence seizures. Next, chronic administration was investigated following an 8 h phase delay. Both experiments were done in symptomatic male WAG/Rij rats, a strain endowed with hundreds spike-wave discharges electroencephalographic (SWDs) (EEG) and daily. Simultaneous electromyographic (EMG) recordings were made during 3 days in the acute and 11 days in the chronic study. Agomelatine showed neither an effect on sleep-wake parameters in the acute study, nor affected the SWDs and reentrainment process in the chronic study. Sleep-wake and SWDs rhythms were advanced immediately as a result of the 8 h delay of the light phase. The magnitude of an advance and the speed of subsequent re-entrainment were different for various rhythms. Coupling between active wakefulness and deep slow-wave sleep, as well as, SWDs and light slow-wave sleep was observed. A post-shift increase in passive wakefulness and a reduction in deep slow-wave sleep resulted in an aggravation of epileptic activity during the light phase. Different speed of re-entrainment and coupling between various rhythms suggest that SWDs and light slow-wave sleep are controlled 139 by common circadian mechanism distinct from that for active wakefulness and deep slow-wave sleep. The increase in the number of seizures after the phase shift may be of significant importance for people with epilepsy planning long trans-meridian flight. Keywords: absence epilepsy, WAG/Rij rats, sleep-wake states, circadian desynchronization, phase delay 1. Introduction Rhythmic occurrence of mammalian physiological and behavioral processes results from daily entrainment of the circadian timing system to environmental changes (Zeitgebers) (Pittendrigh, 1981). True entrainment, a stable phase relationship between the rhythm and the Zeitgeber cycle, is crucial for organism’s adaptation, survival and well-being (Dodd et al., 2005). Misalignments caused by rapid shifts in the timing of the light-dark cycle, lead to circadian desynchronization, negative consequences of which were described in people crossing more than two time zones with a jet, shift workers and also in animal models (Arendt and Marks, 1982; Winget at al., 1984; Lancel and Kerkhof, 1989; Knutsson, 2013 ). Re-synchronization after the shift needs considerable amount of time, different for various functions and rhythms (Yamazaki et al., 2000; Lemmer et al., 2002; Nagano et al., 2003). One of the most popular coping strategies for circadian disturbances is the usage of exogenous melatonin; it has been found beneficial in accelerating a re-entrainment process and therefore ameliorating symptoms of jet lag syndrome both in human and animals (Illnerova et al., 1989; Golombek and Cardinali, 1993; Sharma et al., 1999). The consequences of internal desynchronization caused by the transfer to constant dim light condition and by an 8 h phase delay in 140 the light-dark cycle in WAG/Rij rats, a well-known, validated, genetic animal model of childhood absence epilepsy (CAE) (Depaulis and van Luijtelaar, 2006) have been described. The occurrence of absence seizures, marked by the presence of short-lasting, bilateral and synchronous spike-wave discharges (SWDs) in the electroencephalogram (EEG), is organized into a 24 h rhythm with a maximum taking place during the dark phase and minimum occurring at the beginning of the light phase of 12:12 light-dark cycle (van Luijtelaar and Coenen, 1988). In the absence of entrainment (constant condition of dim light and the phase-shift mentioned above), the rhythm of SWDs was found to desynchronize from the rhythm of motor activity, to which it was tightly coupled under standard laboratory conditions (Smyk et al. 2011; Smyk et al. 2012). Opposite to a gradual, stable re-entrainment of the motor activity to the shifted photoperiod which was completed after 6 days, re-synchronization of the rhythm of SWDs was longer and irregular. Moreover in both conditions (dim light and after the phase shift), the lack of entrainment resulted in an enhancement of epileptic activity (Smyk et al. 2011; Smyk et al. 2012). Considering a close relationship between the occurrence of absence seizures and sleep-wake states (Drinkenburg et al., 1991), we aimed to establish to which sleep-wake states rhythm, if any, SWDs is coupled during re-synchronization process. Agomelatine is a newly introduced antidepressant with chronobiotic properties. This melatonin naphthalene analogue is a potent agonist of melatonin receptors subtypes 1 and 2 (MT1, MT2) and antagonist of serotonin receptor subtype 2 (5-HT2C) (de Bodinat et al., 2010). Antidepressant action of agomelatine has been shown both in animal studies and clinical trials (Loo et al., 2002; Papp et al., 2003; Bourin et al., 2004; Bertaina-Anglade et al., 2006; Goodwin et al., 2009). Chronobiotic properties of the drug have been reported in rats kept in constant darkness, in which activity rhythm entrained to 141 daily administration of agomelatine coinciding with the activity onset (Martinet et al., 1996). Phase-advancing effect of agomelatine, similar to this obtained for melatonin, in an 8 h phase advance paradigm resembling jet-lag were described (Redman et al., 1995). The compound reinstates circadian rhythms in aged rodents and normalizes disrupted sleep-wake cycle in trypanosome-infected rats (Koster-van Hoffer et al., 1993; Grassi-Zucconi et al., 1996; van Reeth et al., 2001). The second goal of the present study was to investigate an effect of chronic administration of various doses of agomelatine on the course of re-entrainment of the rhythms of SWDs and sleep-wake states to the 8 h phase delay. Melatonin affects epileptic activity in various seizure and epilepsy models (Lapin et al., 1998; Musshoff and Speckmann, 2003; Yildirim and Marangoz, 2006); also agomelatine showed anticonvulsant activity in PTZ induced seizures and in the pilocarpine induced status epilepticus model of temporal lobe epilepsy (Aguiar et al. 2012). Melatonin and agomelatine may alter sleep-wake architecture in the rat (Fisher et al., 2008; Descamps et al., 2009). Considering the potential hypnotic and antiepileptic effect of agomelatine, our chronic experiment was preceded by an acute study, which allowed to entangle the putative effects of agomelatine on sleep and epilepsy and the effects of the light phase delay on re-entrainment. Here the effect of a single administration of various doses of agomelatine on the number and mean duration of SWDs, on sleepwake states and circadian parameters of the rhythms of SWDs and sleep-wake states was investigated. 142 2. Materials and methods 2.1. Animals 32 male, adult (8 months of age) WAG/Rij rats purchased from Harlan (Harlan, the Netherlands), 315 g ± 12g of weight, were used. Animals were singly-housed in individually ventilated Plexiglas cages (25 x 22 x 18 cm) in a sound-attenuated room and controlled environmental conditions (temperature: 22 ± 2 C, humidity: 60%, 12:12 light-dark cycle, white lights on at 5 am off at 5 pm , light intensity: ≈ 100 lux) throughout the experiment as well as during recovery period after the surgery. The exception was the shifted LD cycle after the phase delay (see chronic study), then the lights were on 1 pm and off at 1 am. Standard laboratory chow and tap water were available ad libitum. All experimental procedures were approved by the animal use and care committee of Janssen Research and Development, Pharmaceutical Companies of Johnson & Johnson and the local ethical committee. 2.2. Surgery Animals underwent stereotactic surgery under deep isoflurane anesthesia. Seven stainless steel fixing screws (diameter 1 mm) were inserted bilaterally in the left and right hemisphere along the anteroposterior axes in the following coordinates: AP +2 mm, L +/– 2 mm; AP -2 mm, L +/-2 mm and AP -6.6 mm , L +/- 2 mm from Bregma; and referenced to the same ground electrode place midline above of the cerebellum. The incisor bar was –5 mm under the center of the ear bar, according to the stereotactic atlas of Paxinos and Watson. In addition, stainless steel wires electrodes were placed in the muscles of the neck to record EMG activity. Electrodes (stainless steel wire, 143 7N51465T5TLT, 51/46 Teflon Bilaney, Germany) were connected to a pin (Future Electronics: 0672-2-15-15-30-27-10-0) with a small insert (track pins; Dataflex: TRP-1558-0000) and were fit into a 10-holes connector then the whole assembly was fixed with dental cement to the cranium. Rats were allowed to recover for 10 days after surgery. 2.3. EEG and EMG recordings Rats, while staying in their home cages, were placed in soundattenuated, individually ventilated recording boxes and connected by a cable to rotating swivels, allowing free movement during recording procedures. Rats were allowed to habituate to the recording set-up for 24hr preceding experiments. Continuous EEG and EMG signals were acquired at 2 kHz sample rate with an input range of +/- 500 mV through a Biosemi ActiveTwo system (Biosemi, Amsterdam, the Netherlands) referenced to the CMS-DRL ground (common mode reference for online data acquisition and impedance measures, which is a feedback loop driving the average potential across the montage close to the amplifier zero). The signals were amplified and analogue band-pass filtered between 1 and 100 Hz and were digitized with 24 bit resolution. 2.4. Pharmacological treatment Saline, agomelatine (Sigma-Aldrich, Belgium) in doses of 2.5, 5 and 10 mg/kg dissolved in 20% cyclodextrin (Sigma-Aldrich, Belgium) and 20% cyclodextrin alone were administrated orally 1 h before the onset of darkness. 144 2.5. Experimental design The acute study consisted of 3 experimental days. Each day consisted of 24 h of simultaneous and continuous EEG and EMG recordings. Recordings started at the last hour of the light phase during which injections or sham administration took place, then they were continued by 12 h of the dark phase and 11 h of the light phase. Animals were randomly assigned to 4 groups (n=8 rats each): control group, 2.5, 5 and 10 mg/kg group of agomelatine, respectively. On the first, baseline day, all rats received saline. On the second, treatment day, animals received agomelatine in 3 doses: 2.5, 5 and 10 mg/kg, rats of the control group were injected with vehicle. On the third, posttreatment day, oral administration of a compound was mimicked by picking up animals from their cages without usage of feeding tubes and pharmaceutical agents. The chronic study consisted of 11 experimental days and 1 period of extended darkness. Recording schedule was the same as in the acute study. Again, rats were randomly assigned to 4 groups (n=8 rats each): control group, 2.5, 5 and 10 mg/kg of agomelatine, respectively. On the first baseline day, all animals received saline (oral administration) 1 h before the onset of darkness. After that day, an 8 h phase shift in the light-dark cycle was introduced by extending the subsequent dark period from 12 to 20 hr. Recordings were continued for 10 consecutive, post-shift days starting from the last hour of the light phase of the shifted photoperiod. Every day, at the last hour of the light phase (1 h before the onset of darkness), animals were given agomelatine, while animals of the control group received vehicle. Due to a crash of the recording system, the data of the second post-shift day were lost and are missing from the analysis. 145 2.6. Data analysis Five sleep-wake states were automatically identified in 2 sec epochs: active wakefulness, passive wakefulness, light and deep slowwave sleep and rapid-eye-movement sleep (REM), based on EEG, EMG and body movements (PIR detection) following validated criteria (Ahnaou and Drinkenburg, 2011). The number and duration of SWDs across 24 h was scored automatically based on the EEG signal from a frontal electrode against reference according to well-established criteria, next they were verified visually based on criteria published elsewhere (van Luijtelaar and Coenen, 1986; Ovchinnikov et al., 2010). Data from 24 h days of both acute and chronic experiment: time spent in one of the five sleep-wake states (expressed in min.), number and mean duration of SWDs (duration expressed in sec.) were grouped into 1 h bin. Each hour of baseline day was compared with corresponding hours of the treatment and post-treatment days in the acute, and 10 post-shift days in the chronic study, respectively. To estimate circadian parameter acrophase of the rhythms of sleep-wake states and SWDs, data were grouped into 6 min bin, as previously described (Smyk et al., 2012). Cosine wave of a fixed period length of 24 h was fit to each of 3 days of the acute study, baseline and post-shift days of the chronic study (program Cosinor available at http://www.circadian.org/softwar.html). The phase relationship between different rhythms during baseline and post-shift days of the chronic study was determined as a difference between their acrophases (in degrees) on a particular experimental day. Significant difference from the baseline value of the phase relationship between rhythms during post-shift days was interpreted as desynchronization between rhythms. 146 Additionally, the period of extended darkness (8 h before the new light onset) of the chronic study was analyzed. Each hour of the total time of particular sleep-wake states and number of SWDs in the extended darkness was compared with corresponding hours of the beginning of the light and the dark phase of the baseline day. 2.7. Statistical analysis Statistical analyses were done in Statistica (StatSoft, Inc., Tulsa, Oklahoma, USA). Repeated measures analysis of variance (ANOVA) were used to estimate dose (4 levels), hour (24 levels) and day (3 levels: baseline, treatment and post-treatment day) effect of agomelatine treatment, on total duration of the five sleep-wake states, mean duration and number of SWDs in the acute study, and dose (4 levels), phase (light and dark) and day (10 levels) effect of the phase shift and agomelatine treatment in the chronic experiment. Repeated measures ANOVA were also used to estimate the dose and day effect of agomelatine treatment on circadian parameters of the rhythms of sleep-wake states and SWDs in both studies as well as to compare the period of extended darkness, the beginning of the light and the dark phase and phase relationship between rhythms. Bonferroni’s test was used as post-hoc test. Differences were considered to be significant at a p level < 0.05. 3. Results 3.1. Acute study In all sleep-wake states investigated, an hour effect was observed (active wakefulness: F = 235.32, passive wakefulness: F = 37.53, light slow-wave sleep: F = 385.21, deep slow-wave sleep: F = 147 241.82, REM sleep: F = 72.98 and SWDs: F = 53.864, all df = 23, 644, all p < 0.05). In all sleep-wake states (except light slow-wave sleep) and number of SWDs, a day effect was found (active wakefulness: F = 15.48, passive wakefulness: F = 9.96, deep slow-wave sleep: F = 14.11, REM sleep: F = 30.32 and SWDs: F = 9.76, all df = 2, 56, all p < 0.05). Post-hoc tests following the day effect indicated a small increase in passive wakefulness on the treatment day in comparison with the baseline and the post-treatment day (5.95 ± 0.90 min vs 5.62 ± 0.86 min and 5.95 ± 0.81 min, treatment vs baseline and posttreatment day, respectively, all p <0.05) and a small elevation of REM sleep, both on treatment and post-treatment day (7.41 ± 0.74 and 7.81 ± 0.82 min vs 7.17 ± 0.79 min, treatment and post-treatment vs baseline day, respectively, all p < 0.05). A decrease on the treatment day was found for deep slow-wave sleep and number of SWDs (15.31 ± 0.70 min vs 15.81 ± 0.74 and 15.90 ± 0.80 min, treatment vs baseline and post-treatment day, respectively for deep slow-wave sleep, all p < 0.05; 15.01 ± 2.31 vs 17.09 ± 2.32 and 16.90 ± 2.95, treatment vs baseline and post-treatment day, respectively for the number of SWDs), while active wakefulness last shorter only on posttreatment day (18.76 ± 1.19 min vs 19.46 ± 1.15 and 19.78 ±1.31 min, post-treatment vs baseline and treatment day, respectively). A significant day x hour interaction was found for all variables investigated (active wakefulness: F = 5.18, passive wakefulness: F = 4.80, light slow-wave sleep: F = 4.93, deep slow-wave sleep: F = 4.70, REM sleep: F = 7.57 and SWDs: F = 8.18, all df’s = 46, 1288; all p < 0.05). Post-hoc comparison between baseline and treatment day revealed significant differences for single hours only: 6th hour after administration of agomelatine for active wakefulness (38.06 ± 1.26 min vs 30.73 ± 1.19 min, treatment vs baseline day, p < 0.05), 12th for passive wakefulness (7.85 ± 0.31 min vs 5.46 ± 0.31 min, treatment vs baseline day, p < 0.05), 1st, 6th and 24th for light slow148 wave sleep (9.36 ± 0.36 min vs 11.72 ± 0.41; 2.78 ± 0.25 min vs 5.32 ± 0.29 min; 14.51 ± 0.38 min vs 10.30 ± 0.34 min, treatment vs baseline day for 1st, 6th and 24th hour, respectively, all p < 0.05), 24th for deep-slow wave sleep (15.67 ± 0.40 min vs 10.61 ± 0.58 min, treatment vs baseline day, p < 0.05) and 2nd hour for REM sleep (10.43 ± 0.56 min vs 13.45 ± 0.52 min, treatment vs baseline day, p < 0.05). However, the number of SWDs was decreased for 4 consecutive hours (4th, 6th – 8th) on the treatment day compared to baseline and post-treatment day (4th hour: 25.56 ± 2.13 vs 15.94 ± 1.17 treatment vs baseline day, p < 0.05; 6th hour: 14.65 ± 1.50 vs 29.16 ± 1.71 and 30.87 ± 2.92, treatment vs baseline and posttreatment day, all p < 0.05; 7th hour: 14.91 ± 1.35 vs 31.28 ± 2.53 and 32.91 ± 2.11, treatment vs baseline and post-treatment day, all p < 0.05; 18.34 ± 1.81 vs 34.09 ± 2.26 and 29.06 ± 1.98, treatment vs baseline and post-treatment day, all p < 0.05). There was neither a dose nor a dose and day interaction, nor between day, dose and hours, as could have been predicted in case agomelatine affected sleep-wake states or the number of SWD in a couple of hours on the treatment day. The data on SWD occurrence across the 24h period in all the three days are presented in Figure 1. Analysis of acrophase of the rhythms of sleep-wake states and SWDs revealed neither a dose effect of agomelatine nor an interaction between dose and day. Significant results regarded day effects only (active wakefulness: F = 29. 80, df = 2, 56, p < 0.05; deep slow-wave sleep: F = 7.09, df = 2, 56, p < 0.05; REM sleep: F = 21.56, df = 2, 56; SWDs = 14.78, df = 2, 56, p < 0.05). In all cases analyzed, with one exception of light slow-wave sleep, which was not affected by agomelatine at all, acrophase of the rhythms was slightly advanced on the treatment day in comparison to baseline (active wakefulness: 84.72 ± 1.19 vs 91.62 ± 1.15 degrees; deep slow-wave sleep: 63.04 ± 4.37 vs 84.04 ± 2.77 degrees; REM sleep: 309.19 ± 2.58 vs 329.09 149 ± 2.34 degrees; SWDs: 63.04 ± 4.37 vs 84.042.78 degrees, treatment day vs baseline, all p < 0.05). The effect noticed at the treatment day was still present at the post-treatment day since acrophases of the rhythms of active wakefulness, deep slow-wave sleep, REM sleep and SWDs were still advanced (all p < 0.05). The acrophase of active wakefulness was even more advanced than on the treatment day (91.62 ± 1.15 vs 84.72 ± 1.19 vs 80.22 ± 1.18 degrees, baseline vs treatment vs post-treatment day, all p < 0.05). Figure 1. Mean and SEM of the number of SWDs across 24 h on (A) baseline, (B) agomelatine treatment and (C) post-treatment day of the acute study. Doses: 2.5 mg/kg (n = 8) – green line, 5 mg/kg (n = 8) – blue line, 10 mg/kg (n = 8) – red line, control group (n = 8, 20% cyclodextrin) – black line. Resolution of the data: 1 h. Mean and SEM (n = 32) of the number of SWDs across 24 h during all experimental days of the acute study (D). Baseline day – black line, agomelatine treatment day – dark grey line, post-treatment day – light grey line. The dark phase of the 12:12 light-dark cycle is marked by the grey rectangle 150 3.2. Chronic study Circadian rhythms in all sleep-wake states and in the occurrence of SWDs were found on the baseline day. Rats spent more time in active wakefulness during the dark period of 12:12 light-dark cycle (337.95 ± 6.31 min vs 129.34 ± 4.20 min, p < 0.05), they had also more absence seizures (295.12 ± 31.43 vs 102.38 ±11.72, p < 0.05). The light phase was dominated by slow-wave and REM sleep (light slow-wave sleep: 152.94 ± 2.22 vs 65.71 ± 1.53 min, deep slow-wave sleep: 253.57 ± 3.58 vs 131.18 ± 2.94 min, REM sleep: 93.73 ± 2.62 vs 75.61 ± 2.20 min, light vs dark, respectively, all p < 0.05). The phase-related difference in the total duration of passive wakefulness was not significant (72.30 ± 2.64 vs 67.73 ±2.21, dark vs light, p > 0.05). Repeated measures ANOVA for the total time spent in active wakefulness in the light and the dark phase over all recorded days (baseline and 9 post-shift days) showed a significant day (F = 10.28, df = 9, 504, p < 0.05), phase (F = 743.91, df = 1, 56, p < 0.05) and an interaction (day x phase) (F = 243.24, df = 9, 504, p < 0.05) effect. Significant dose effect and interactions with dose such as dose x phase, dose x day and dose x day x phase were not found. Post-hoc comparison between days showed that the total duration of active wakefulness was decreased at 1st, 3rd and 4th day after the phase shift (222.13 ± 4.13 min, 216.07 ± 3.71 min and 222.81 ± 3.79 min vs 233.64 ± 4.62 min, all p < 0.05). The phase-shift caused a decrease in the duration of active wakefulness in the dark phase accompanied by an increase in the light phase which last from 1st to 7th post-shift day. Data are presented in Figure 2. Repeated measures ANOVA for the total time spent in passive wakefulness in the light and the dark phase over all recorded days showed a significant day (F = 6.63, df = 9,504, p < 0.05) and an 151 interaction (day x phase) (F = 54.51 df = 9,504, p < 0.05) effect. Neither phase and dose effects, nor interactions with dose were found. Total duration of passive wakefulness during almost all post-shift days (exceptions: 5th, 6th and 8th day) was elevated as revealed by posthoc tests (all p’s < 0.05). Also phase-dependent changes were found. A robust increase in the duration of passive wakefulness was observed in the light phase from 1st to 4th post-shift day (significant increase was still observed on 5th and 7th post-shift day, all p’s < 0.05). A nocturnal level was decreased on the 1st and 3rd post-shift day and also during last 3 days of the experiment (Figure 2). Dose effects and interactions: dose x phase, dose x day and dose x day x phase were not found. Significant day (F = 3.18, df = 9, 504, p < 0.05), phase (F = 1423.45, df = 1, 56, p < 0.05) and day x phase (12.66, df = 9, 504, p < 0.05) effect was found for total duration of light slow-wave sleep over all experimental days. Changes of the total time spent in light slow-wave sleep in the light and the dark phase were less pronounced as in comparison to active and passive wakefulness. Both diurnal and nocturnal level of light slow wave sleep was increased and reduced, respectively only for 2 days after the phase shift. Data presented in Figure 2. Analysis of variance of total time spent in deep slow-wave sleep showed significant phase (F = 359.56, df = 1, 56, p < 0.05) and day x phase interaction effect (F = 472.70, df = 9,504, p < 0.05). Neither day nor dose effect were found (F = 1.65, df = 9, 504 and F = 0.65, df = 3, 56, respectively, all p’s > 0.05), nor interactions with dose. Total duration of deep slow-wave sleep was significantly elevated in the dark phase during first 5 days after the phase-shift (216.21 ± 2.70 min, 198.03 ± 3.02 min, 176.26 ± 3.19 min, 161.74 ± 3.24 min and 148.85 ± 3.48 min vs 131.18 ± 2.94, 1st, 3rd, 4th, 5th and 6th postshift day vs baseline, all p’s < 0.05) while duration in the light phase 152 was decreased for four days (162.75 ± 2.84 min, 192.85 ± 3.29 min, 209.82 ± 3.32 min and 161.74 ± 3.24 min vs 253.57 ± 3.58, 1st, 3rd, 4th, and 5th post-shift day vs baseline, all p < 0.05). On the first postshift day, typical phase-related difference: longer duration of deep slow-wave sleep during the dark period was reversed (p < 0.05). The restoration took place on 4th post-shift day and stayed significant from that moment onwards (all p < 0.05) (Figure 2). Repeated measures ANOVA for the total time spent in REM sleep in the light and the dark phase over all recorded days showed a significant day (F = 12.71, df = 9,504, p < 0.05), phase (F = 17.35, df = 1, 56, p < 0.05) and an interaction (day x phase) (F = 28.87 df = 9,504, p < 0.05) effect. No dose effect was found, nor an interaction with dose. Post-hoc comparison revealed that the duration of REM sleep was increased starting from the 1st to 6th post-shift day (all p < 0.05), while between-phases comparison showed that this is a result of elevated level of REM sleep in the dark phase, since large significant differences between baseline and post-shift days were found only during the darkness (1st – 7th post-shift day, all p < 0.05). 153 Figure 2. Mean and SEM (n = 32) of the (A) total duration of sleep-wake states and (B) total duration of sleep-wake states in the light and the dark phase of 154 the 12:12 light-dark cycle before and after an 8 h light phase delay. AW – active wakefulness, yellow line; PW – passive wakefulness, red line; SWS L – light slow-wave sleep, green line, SWS D – deep slow-wave sleep, dark green line, REM – REM sleep, blue line. The light phase is marked by a dotted line, the dark phase is marked by a solid line. * p < 0.05, baseline vs post-shift days, ANOVA for repeated measures design, Bonferroni post-hoc test Significant phase (F = 15.52, df = 1, 7, p < 0.05) and day x phase interaction (F = 21.04, df = 9, 36, p < 0.05) for the number of SWDs, day (F = 3.87, df = 9, 36, p < 0.05) and day x phase interaction (F = 3.28, df = 9, 36, p < 0.05) effect for mean duration of SWDs were found. Post-hoc tests revealed that in the light phase rats had more seizures on first 4 post-shift days in comparison to baseline, while during same number of post-shift days in the dark phase absences were less numerous (all p < 0.05). Similarly to deep slow-wave sleep a phase-related redistribution was seen on the 1st post-shift day, however it was not significant (data presented in Figure 3). The mean duration of SWDs was increased from 3rd to 4th and post-shift days (p < 0.05), with the increase in the light phase (first 3 post-shift days, all p < 0.05) as the main contributor to this effect (Figure 3). There was no dose effect or interactions with dose on number and mean duration of SWDs. Circadian rhythmicity was detected in all sleep-wake states and the occurrence of SWDs on the baseline day. The acrophase of active wakefulness and SWDs was found to occur in the dark phase, while light slow-wave sleep, deep slow-wave sleep and REM sleep peaked in the light phase. The acrophase of passive wakefulness rhythm was found around a light/dark transition (at the end of the light phase in some rats while at the beginning of the dark phase in others), therefore, not the acrophase per se but the difference in degrees between the baseline value and each consecutive day was analyzed. 155 Figure 3. Mean and SEM (n = 8) of the total number and mean duration of SWDs (A) and the total number and mean duration of SWDs in the light and the dark phase of the 12:12 light-dark cycle (B) before and after an 8 h light phase delay. The light phase is marked by a dotted line, the dark phase is marked by a solid line. * p < 0.05, baseline vs post-shift days, ANOVA for repeated measures design, Bonferroni post-hoc test In general, dose effect of agomelatine and its interactions were not found in acrophases of the rhythms investigated. To avoid that the outcomes of circadian analyses were overpowered by the large number of animals (n = 32), only the data from the control group (n = 8) were used to illustrate the process of re-entrainment of various rhythms to the phase shift. Analysis of variance (ANOVA, repeated measures design) for the rhythms of sleep-wake states and SWDs over all experimental days (baseline and 9 post-shift days) showed that the position of acrophase changed over days in all states and SWDs with an exception of passive wakefulness (active wakefulness: F = 118.44, light slow-wave sleep: F = 67.88, deep slow-wave sleep: F = 99.06, 156 REM sleep: 13.92, SWDs: F = 10.08, all df = 9, 63, all p < 0.05). 8 h phase shift in the 12:12 light dark cycle caused an advanced of all acrophases of 72.95 ± 25.07 degrees (active wakefulness: from 81.50 ± 2.41 to 12.38 ± 2.90 degrees, light slow-wave sleep: from 274.50 ± 1.94 to 228.25 ± 2.28 degrees, deep slow-wave sleep: from 226.87 ± 2.96 to 156.12 ± 1.88 degrees, REM sleep: from 315.75 ± 4.03 to 180.12 ± 11.43 degrees and SWDs: from 65.25 ± 6.44 to 19.25 ± 3.65 degrees). Phase-shift magnitude (the difference between the position of acrophase between the baseline and the 1st phase-shift day) was different for the different sleep-wake states (one-way ANOVA with sleep-wake states-SWD as between subjects factor: F = 25.95, df = 4, 35; p < 0.05). Post-hoc tests revealed that although the acrophase of REM sleep was affected the most, REM sleep accomplished the largest daily shifts (REM sleep vs remaining sleepwake states and SWDs, all p < 0.05) and was the fastest to re-entrain: the acrophase of the REM rhythm was no longer different from baseline level on the 6th post-shift day. Light slow-wave sleep and SWDs reached the baseline values on the 7th day after the shift. Active wakefulness and deep slow-wave sleep showed similar speeds of reentrainment. However, their re-synchronization took one day longer and was accomplished on the 8th post-shift day. Data are presented in Figure 4. During the re-synchronization process, circadian rhythmicity, determined by a significant fit of the cosine curve to the data, was present in all animals, during all experimental days after the phaseshift in following variables: active wakefulness, light and deep slowwave sleep. Circadian rhythms of the number of SWDs, passive wakefulness, and REM sleep could not be well fitted by a cosine and therefore it is suggested that these rhythms showed a poor temporarily organization. Two latter states showed the greatest abnormalities: even on the 10th post-shift day, the expression of passive wakefulness and REM sleep in some rats was not rhythmic. 157 Analysis of variance (ANOVA, repeated measures design) for the phase relationships between rhythms of sleep-wake states and SWDs over all experimental days (baseline and 9 post-shift days) showed significant day effect in all variables investigated (active wakefulness light slow-wave sleep: F = 10.06, active wakefulness - deep slow-wave sleep: F = 2.75, active wakefulness - REM sleep: F = 4.56; light slowwave sleep - deep slow-wave sleep: F = 8.68, light slow-wave sleep SWDs: F = 8.68, deep slow-wave sleep - REM sleep: F = 8.27, deep slow-wave sleep - SWDs: F = 5.59; REM sleep - SWDs: F = 6.34; all df = 9, 63, all p < 0.05) with two exceptions: relationship between active wakefulness and SWDs, where it just failed to reach significance (F = 2.03, df = 9, 63; p = 0.05) and relationship between light slowwave and REM sleep (F = 1.63, df = 9, 63; p > 0.05). Post-hoc comparison revealed that the phase relationship between activewakefulness and light slow-wave sleep changed across the days after the phase shift: from 1st to 5th, 7th and 9th post-shift day. Difference between acrophases of light and deep slow-wave sleep was changed for 3 days after the shift and additionally on the 5th post-shift day, while alterations in phase relationship between active wakefulness and REM sleep, light and deep slow-wave sleep, deep slow-wave sleep and SWDs, REM sleep and SWDs lasted for 3 days after the phase shift. Post-hoc comparison revealed no changes over days in a phase relationship between active wakefulness and deep slow-wave sleep, as well as light slow-wave sleep and SWDs. 158 Figure 4. Mean and SEM (n = 8) of the total duration of sleep-wake states and number of SWDs during the baseline and all post-shift days (upper panel). Data resolution: 1 h. Mean and SEM (n = 8) of the acrophase of vigilance and 159 SWDs rhythms during the baseline and after an 8 h light phase delay (lower panel). Active wakefulness – orange line, passive wakefulness – red line, light slow-wave sleep – green line, deep slow-wave sleep – dark green line, REM sleep – blue line, SWDs – black line. The dark phase of the 12:12 light-dark cycle is marked by the grey rectangle. * p < 0.05, baseline vs post-shift days, ANOVA for repeated measures design, Bonferroni post-hoc test Analysis of the period of extended darkness for all animals revealed a phase, an hour and an hour x phase interaction effect. Posthoc comparison between phases showed that the total time spent in active and passive wakefulness, deep slow-wave sleep and also number of SWDs recorded during extended darkness resembles the light phase of the baseline and is significantly different from the dark phase of the baseline (all p < 0.05, data presented in Figure 5). The response of light slow-wave and REM sleep to an unusual extension of the dark phase was quite different. Total time spent in light slow-wave sleep during the extended dark period was greater than during the dark phase (8.65 ± 0.16 vs 5.28 ± 0.16 min, p < 0.05) but smaller than during the light phase (8.65 ± 0.16 vs 12.63 ± 0.16 min, p < 0.05). Total time of REM sleep during the extended darkness was significantly longer in comparison to the light and dark baseline periods (10.29 ± 0.20 vs 7.01 ± 0.20 and 5.84 ± 0.16 min, extended darkness vs light and the dark phase, respectively, all p < 0.05). Detailed, hour by hour comparisons between phases are presented in Figure 5. 160 161 Figure 5. Mean and SEM (n = 32) of the total duration of active wakefulness (A), passive wakefulness (B), light slow-wave sleep (C), deep slow-wave sleep (D), REM sleep (E) and the number of SWDs (F) (upper panels) during the 8 first hours of the baseline light phase, the 8 first hours of the baseline dark phase and 8 hours of the extended darkness on the phase-shift day. Mean and SEM (n = 32) of the total duration of vigilance states and the number of SWDs in particular hours of the phases (lower panels). The light phase – grey line/block, the dark phase – black line/block, extended dark period – dark green line/block. *, # p < 0.05 extended darkness vs dark phase, extended darkness vs light phase, respectively, ANOVA for repeated measures design, Bonferroni post-hoc test 4. Discussion Re-synchronization of various sleep-wake states and absence seizures to an 8 h light phase delay was investigated in a validated animal model of CAE. Main questions addressed by the study were: how fast different sleep-wake states and SWDs re-synchronize to a shifted photoperiod and whether, given a well-established relationship between absences and sleep-wake states, the rhythm of SWDs is coupled to any of the other rhythms. Second goal was to evaluate the effect of agomelatine, an antidepressant drug with chronobiotic properties, on the duration of sleep-wake states and the number of SWDs during stable entrainment (acute study) and after the phase shift (chronic study). Different speeds of the re-entrainment of sleep-wake states and SWDs rhythms, coupling between various rhythms during the resynchronization process, different effect of the prolonged darkness on the rhythms and a lack of effects of agomelatine neither in the acute, nor in the chronic experiment comprise the main findings of the present study. 162 4.1. Course of re-entrainment of the sleep-wake states and SWDs rhythms Physiological consequences of circadian misalignments produced by rapid phase shifts in the photoperiod are well documented both in humans as well as in animal models (Winget et al., 1984; Lancel and Kerkhof, 1989). The number of hours and the direction of the shift (advance vs. delay) are critical for a speed of subsequent reentrainment (Wever, 1966). In general, phase delay adjustments corresponding to westward travels are accomplished faster than phase advances, an equivalent of eastward travels (Lemmer et al., 2002; Murphy et al., 1998; Reddy et al., 2002; Yan, 2011). Within the limits of one direction of the shift, distinct rhythms may re-entrain with different speed. An internal desynchronization between circadian oscillators and rhythms is quite common (Nagano et al., 2003; Lee et al., 2009). In the present study, an 8 h light phase delay was produced by extending the dark period from 12 to 20 h. As a result, all rhythms investigated were found to be advanced with respect to the new photoperiod. Changes in total duration and phase-related distribution of all sleep-wake states, number and mean duration of SWDs were found. 4.1.1. Active wakefulness Animal studies investigating re-entrainment of all sleep-wake states after the phase shift are scarce. Usually only active wakefulness or general activity, measured by means of passive infrared sensors or telemetrically, or wheel running activity across days are reported. In general, the time required for re-entrainment of these rhythms (in days) is smaller than the number of hours by which the phase was delayed (Takamure et al., 1991; Yamazaki et al., 2000). In rodents, 163 the re-entrainment process was accompanied by a redistribution of activity between the new light and the dark period (Kopp et al., 1998; Nagano et al., 2003). In our previous study in which the same phase shift paradigm in WAG/Rij rats was used, re-synchronization of general motor activity was complete on the 7th post-shift day, which is one day earlier in comparison to the present result. A similar redistribution of activity was observed, however, the decrease of the nocturnal level lasted one day longer (8 days), while the diurnal level was increased for 4 post-shift days only (Smyk et al., 2012). Given the lack of any effect of agomelatine in both the acute and chronic study, differences between the present and our previous studies in which rats were not injected, might be attributed to the additional manipulations necessary for the administration of the solvent or drug. The injections, handling and transient arousals repeated daily during all 10 post-shift days might be considered as another, non-photic stimulus for the circadian timing system. The presence of such stimulus might have affected the re-synchronization process to some degree. The possibility cannot excluded that factors such as handing and injections are stressful, however, in habituated animals these procedures are generally not considered as stressors. 4.1.2. Passive wakefulness A decrease in the total amount of active wakefulness was followed by an increase in passive wakefulness occurring immediately after the shift and lasting the same number of days. The decrease of activity or wake state in general has been described previously. A study of Beaumont and coworkers (2004) might be linked to some extent to the decrease in activity reported by us; they investigated effects of slow-release caffeine and melatonin on sleep and daytime sleepiness after a seven-time zone eastward flight. Subjects receiving placebo 164 were significantly drowsier until 6th day after the flight in comparison to baseline as measured by modified EEG sleep latency test (Beaumont et al., 2004). 4.1.3. Light and deep slow-wave sleep The total duration and the phase-related distribution of light slow-wave sleep were hardly affected by the phase shift. A similar effect regarding total sleep duration was seen also for deep slow-wave sleep. However and in contrast, the phase-related changes of deep slow wave sleep were much more pronounced and long-lasting. Similar results with respect to the amount and distribution of slow-wave sleep, however, in a phase advance paradigm, were obtained by Sei and coworkers (Sei et al., 1992, 1994). The less adverse effect of the phase delay on slow wave sleep in the present study may account for the lack of changes in total duration of slow-wave sleep during post-shift days. Moreover, the study cited did not distinguished between the two stages of slow-wave sleep. Human polysomnographic studies revealed that an 8 h phase delay had a small effect on slow-wave sleep (only a trend towards smaller amounts of slow-wave sleep on the first post-shift day was observed, the distribution of slow-wave sleep remained unchanged), while the phase advance resulted in an increase in the amount of slow-wave sleep up to 5 days (Buxton et al., 2000; Beaumont et al., 2004). 4.1.4. REM sleep REM sleep was elevated for 6 days, this was in contrast to both light and deep slow-wave sleep. The increase in REM sleep was restricted to the dark phase. The increase in REM sleep after a phase shift was reported previously in a phase advance paradigm in rats, as 165 well as, in mice under chronic circadian disruption protocol, during which animals were exposed weakly to 6 h phase advances of the lightdark cycle (Sei et al., 1992, 1994; Castanon-Cervantes et al., 2010; Brager et al., 2013). In humans, a study in a laboratory controlled 8 h light-dark and sleep-wake cycle phase delay, abnormalities in the distribution of REM sleep up to 3 post-shift days were found (Buxton et al., 2000). 4.1.5. SWDs The phase delay affected also the epileptic activity of the rats: while the total number of SWDs after the phase shift remained unchanged, a phase-related redistribution occurred. SWDs were more prevalent during the light phase and less during the dark phase. SWDs are tightly related to a momentary state of vigilance: passive behavioral states such as passive wakefulness and light slow-wave sleep enhance the occurrence of seizures; active wakefulness, deep slow-wave and REM sleep have an inhibitory effect (Drinkenburg et al., 1991). In the present study, the elevation of the number of SWDs during the light phase is accompanied by the increase in the total duration of passive wakefulness and a marked reduction of deep slowwave sleep. Therefore, a balance between SWDs enhancing (passive wakefulness, light slow-wave sleep) and inhibiting (deep slow-wave sleep) brain states is shifted towards favorable conditions for SWDs to occur. Similar results were obtained in our previous study, in which WAG/Rij rats were exposed to the same light-dark manipulation however, without pharmacological intervention (Smyk et al., 2012). Both studies showed the increase in the occurrence of seizures in the light phase, during which SWDs are usually less frequent. Different types of seizures e.g. temporal lobe seizures, frontal lobe seizures, 166 tend to occur rhythmically at the particular time of a day (Quigg et al., 1998; Pavlova et al., 2004; Durazzo et al., 2008; Hofstra et al., 2009). As a result of circadian desynchronization the timing of seizures may change. Hypothetically, temporal lobe seizures which occur usually during the day, after a rapid crossing of considerable number of time zones may occur during the night (Quigg et al., 1998). This may lead to additional sleep disturbances and worsening of jet lag symptoms since convulsive seizures occurring during the night have been shown to cause sleep fragmentation, reduction of particular sleep stages and daytime somnolence (Bazil et al., 2000). Absences, known to be related to sleep-wake states and thus occurring more frequently during drowsiness, sleep and state transitions (Halasz et al., 2002) may increase during the day due to enhanced sleepiness and also during the night due to poor, fragmented sleep. To avoid possible aggravation of epileptic activity patients should adjust the timing of antiepileptic medications or prepare themselves to the new time zone by gradual, everyday shifts before the journey. The possibility that also the dose needs an adaptation, cannot be excluded at forehand. 4.1.6. Internal desynchronization Internal desynchronization after a rapid shift in the photoperiod has been found both in humans and animal models (Wegman et al., 1986; Nagano et al., 2003; Lee et al., 2009). In the present study different speed of re-entrainment of various rhythms and changes in their phase relationships was observed. The acrophase of REM sleep was shifted the most; however, it re-synchronized as first, on the 6th post-shift day. REM sleep propensity is well known to be controlled by the circadian timing system (Czeisler et al., 1980). However, the amplitude of the rhythm is small in comparison to, for example, activity or slow-wave sleep (Borbély, 1975). It is suggested that rhythms with 167 small circadian amplitude accomplished larger acrophase shifts and their rate of re-synchronization is faster (Reinberg et al., 1979; Winget et al., 1984). In agreement with this assumption, the rhythm of active wakefulness and deep slow-wave sleep, both of large circadian amplitude, completed the re-entrainment process the slowest. Their acrophases reached the baseline level on 8th post-shift day. Rhythms of light slow-wave sleep and SWDs, both with average amplitude, completed the shift one day earlier. REM sleep, characterized by the lowest circadian amplitude, re-synchronized the fastest. During an extended dark period preceding the light phase delay, rhythms which re-synchronized together showed similar behavior. Active wakefulness, deep slow-wave sleep but also SWDs rhythm behaved as during corresponding hours of the light phase. Similar results were obtained for general motor activity and the number of SWDs in the previous study (Smyk et al., 2012). Light slow-wave and REM sleep showed different behavior hardly comparable with either the light or the dark phase. Desynchronization between different states of sleep-wake states after the phase shift and under a forced desynchrony protocol has been described previously in rats (Sei et al., 1992, 1994; Cambras et al., 2007; Lee et al., 2009). It is thought to be a consequence of desynchrony between anatomically and functionally distinct subregions of the SCN, to which particular sleep-wake states are coupled to. Circadian regulation of sleep, slow-wave sleep and locomotor activity was found to be associated with the activity of both the ventrolateral and dorsomedial SCN, while circadian regulation of REM sleep dependent merely on the activity of the dorsomedial SCN (Cambras et al., 2007; Lee et al., 2009). In the present study, significant post-shift changes in phase relationship between active wakefulness and REM sleep as well as deep slow-wave and REM sleep were found, which suggests internal desynchronization between these sleep-wake states. 168 On the other hand, difference between acrophases of active wakefulness and deep slow-wave sleep remained unchanged, which might be interpreted as stable coupling between these rhythms also during re-entrainment. The response of active wakefulness and deep slow-wave sleep to the phase shift, their re-entrainment rate and stable phase relationship may suggest that indeed they are governed by the same mechanism distinct from REM sleep, which they uncoupled from. The rhythm of SWDs and light slow-wave sleep shared the same speed of re-synchronization, their phase relationship was stable over the post-shift days. However, the response of the rhythms to the extended period of darkness was quite different. Light slow-wave sleep is one of the most favorable states of vigilance for the occurrence of SWDs (Drinkenburg et al., 1991). Same length of the re-entrainment process may suggest that the rhythms share common circadian mechanism. Different response to the lengthened period of darkness may be related to various level of circadian/exogenous component of the rhythms or differences in the magnitude of masking. 4.2. Lack of effects of agomelatine on states of sleepwake states and SWDs – acute study Neither dose-dependent nor drug effects of agomelatine on sleep-wake states and number of SWDs were found in the acute study, only a day effect. Dosage of agomelatine selected for the present study was based on its circadian synchronizing abilities. According to the study of Martinet and co-workers (1996), dose-dependent effect of agomelatine on a free-running rhythm of running wheel activity was observed between 2.5 to 10 mg/kg, with a dose of 5.7 mg/kg providing true entrainment (Martinet et al., 1996). The highest dose used in the present study (10 mg/kg) was found to induce changes in sleep-wake 169 architecture in a period between 4 to 7 hours after the administration and also during the following day (Descamps et al., 2009). On the other hand, given shortly before the dark onset, agomelatine failed to affect sleep-wake states in nocturnal rodents (Tobler et al., 1994; Huber et al., 1998; Langebartels et al., 2001; Monti et al., 2001). Considering the effect observed during the treatment day, similar changes in the total duration of sleep-wake states were found for REM sleep (increase) and active wakefulness (decrease, but only on the post-treatment day), while decrease in the duration of slow-wave sleep was opposite to changes reported by Descamps et al. (2009). Since neither a dose, nor a drug or interaction with dose was found in the present study, other effects such as day affects cannot be attributed to the action of the compound rather they may have been produced by the vehicle. Cyclodextrin (20%) was used as a vehicle and it was given only on day 2. Cyclodextrins are cyclic oligosaccharides containing 6 to 8 glucose units connected by α-(1, 4) bonds. This particular formation (hydrophilic outside, hydrophobic inside) accounts for their ability to create inclusion compounds with other hydrophobic molecules increasing their stability, solubility and bioavailability and are widely used (Duchene et al., 1986; Davis and Brewster, 2004). Considered as neutral solvent, cyclodextrins were reported to exert neuroactive effect both in vitro and in vivo (Wang et al., 1997; Shu et al., 2004; Pytel et al., 2006). In WAG/Rij rats, hippocampal microinjections of cyclodextrin (45% solution) had a biological relevant effect (it reduced the number of SWDs to 1 h after the administration) (Tolmacheva and van Luijtelaar, 2007). In the present study, the number of SWDs was decreased during the treatment day between 4 to 8 h after administration. Differences in time, during which the effect was manifested may be attributed either to the route of administration: local vs systemic or, considering a slight passage of cyclodextrin through blood-brain barrier (Monnaert et al., 2004), behavioral effects 170 of the administration such as arousal, stomach load, increased thirst may be responsible for the effect observed. 4.3. Lack of effects of agomelatine on sleep-wake states and SWDs – chronic study No drug or dose-dependent effect of agomelatine was found in the chronic study. Daily oral administration of agomelatine did neither affect any of the characteristics of the rhythms investigated, nor the speed of the re-synchronization process. Chronobiotic properties of melatonin and its analogues were found in a phase advance paradigm, in which, physiological rhythms are delayed with respect to the new photoperiod and require to be advanced to reach a stable phaserelationship with the Zeitgeber (Golombek and Cardinali, 1993; van Reeth et al., 1998). In the phase delay design, melatonin was found either non effective or delaying the re-synchronization process (Singaravel et al., 1996). Nonetheless, due to its beneficial effect on the subjective sleep, alertness and performance, melatonin treatment is advised to ameliorate symptoms of jet lag also after westward flights (Deacon and Arendt, 1995; Arendt, 1999). In comparison to the previous study in which general motor activity and the rhythm of SWDs were investigated under the same experimental design, the speed of re-synchronization of both rhythms was slightly different (Smyk et al., 2012). The rhythm of general motor activity re-entrained one day faster than the rhythm of active wakefulness, while the rhythm of SWDs re-entrained one day earlier in the present study. An absence of dose-dependent effect of agomelatine and differences between studies in the speed of entrainment suggest not only that actions accompanying the procedure of peri oral injections (removal from cage, associated handling) might have affected the reentrainment process (like in the study of Hastings et al., 1992), but 171 also obscured the putative positive effects of agomelatine on speed or re-entrainment. Put into other words: the transient arousal produced by agomelatine administration might have masked the effect of the drug on the rhythms. Another possibility might be that doses selected for our study were too small to induce any changes. Martinet and coworkers (1996) reported that a dose of 5.7 mg/kg was effective in providing entrainment (Martinet et al., 1996). However, the experiment was conducted in constant darkness while in the present study rats were kept in 12:12 light-dark cycle before and also after the phase shift. It cannot be excluded that the presence of this powerful Zeitgeber outweighed the effects of agomelatine. Assessment of the level of melatonin during the study would allow more precise explanations. To conclude, the present study demonstrates internal desynchronization between various sleep-wake states and absence seizures after a rapid shift in the timing of the light phase of the photoperiod. Rhythms, thought to be controlled by common circadian mechanism, such as active wakefulness and deep slow-wave sleep showed coupling during the process of re-synchronization. The SWDs rhythm re-entrained together with light slow-wave sleep, a state frequently preceding the occurrence of absence seizures. This may suggest a common mechanism for these functions, distinct from the one governing activity and deep slow-wave sleep. Circadian misalignment caused by the phase shift resulted in an aggravation of epileptic activity especially in the light phase, during which seizures in WAG/Rij rats are sparse. An increased duration of passive wakefulness seems to be responsible for the increase in SWD, since, together with light slow-wave sleep, this sleep-wake state is favorable for the occurrence of SWDs. Treatment with melatonin agonist was found to be ineffective in the acute sleep-wake study, as well in the phase delay paradigm. It had also no effect on epileptic activity. The redistribution 172 of the sleep-wake states across the 24 h during the entrainment process might be of significant importance for people with epilepsy planning transmeridian flights and dealing with consequences of jet lag. 5. Acknowledgements The authors would like to thank Dirk Nuyts for his excellent technical assistance and Dr. Abdallah Ahnaou for fruitful scientific discussions. The authors would like to express the gratitude to Janssen Research & Development, a Division of Janssen Pharmaceutica NV, Beerse, Belgium for supplying facilities to perform the study. 6. References Aguiar CC, Almeida AB, Araújo PV, Vasconcelos GS, Chaves EM, do Vale OC, Macêdo DS, de Sousa FC, Viana GS, Vasconcelos SM. 2012. Anticonvulsant effects of agomelatine in mice. Epilepsy Behav. 24: 324-328. Ahnaou A, Drinkenburg WH. 2011. Neuromedin U(2) receptor signaling mediates alteration of sleep-wake architecture in rats. Neuropeptides.45: 165-174. Arendt J, Marks V. 1982. Physiological changes underlying jet lag. Br Med J (Clin Res Ed). 284: 144–146. Arendt J. 1999. Jet-lag and shift work: (2). Therapeutic use of melatonin. J R Soc Med. 92: 402-405. Bazil CW, Castro LH, Walczak TS. 2000. Reduction of rapid eye movement sleep by diurnal and nocturnal seizures in temporal lobe epilepsy. Arch Neurol. 57: 363368. Beaumont M, Batéjat D, Piérard C, Van Beers P, Denis JB, Coste O, Doireau P, Chauffard F, French J, Lagarde D. 2004. Caffeine or melatonin effects on sleep and sleepiness after rapid eastward transmeridian travel. J Appl Physiol (1985). 96: 50-58. Bertaina-Anglade V, la Rochelle CD, Boyer PA, Mocaër E. 2006. Antidepressant-like effects of agomelatine (S20098) in the learned helplessness model. Behav. Pharmacol. 17: 703–713. 173 Borbély AA, Huston JP, Waser PG. 1975. Control of sleep states in the rat by short lightdark cycles. Brain Res. 95: 89-101. Bourin M, Mocaër E, Porsolt R. 2004. Antidepressantlike activity of S20098 (agomelatine) in the forced swimming test in rodents: involvement of melatonin and serotonin receptors. J. Psychiatry Neurosci. 29: 126–133. Brager AJ, Ehlen JC, Castanon-Cervantes O, Natarajan D, Delisser P, Davidson AJ, Paul KN. 2013. Sleep loss and the inflammatory response in mice under chronic environmental circadian disruption. PLoS One. 8: e63752. Buxton OM, Copinschi G, Van Onderbergen A, Karrison TG, Van Cauter E. 2000. A benzodiazepine hypnotic facilitates adaptation of circadian rhythms and sleepwake homeostasis to an eight hour delay shift simulating westward jet lag. Sleep. 23: 915-927. Cambras T, Weller JR, Anglès-Pujoràs M, Lee ML, Christopher A, Díez-Noguera A, Krueger JM, de la Iglesia HO. 2007. Circadian desynchronization of core body temperature and sleep stages in the rat. Proc Natl Acad Sci U S A. 104: 76347639. Castanon-Cervantes O, Wu M, Ehlen JC, Paul K, Gamble KL, Johnson RL, Besing RC, Menaker M, Gewirtz AT, Davidson AJ. 2010. Dysregulation of inflammatory responses by chronic circadian disruption. J Immunol. 185: 5796-5805. Czeisler CA, Zimmerman JC, Ronda JM, Moore-Ede MC, Weitzman ED. Timing of REM sleep is coupled to the circadian rhythm of body temperature in man. Sleep. 2: 329-346. Davis ME, Brewster ME. 2004. Cyclodextrin-based pharmaceutics: past, present and future. Nat Rev Drug Discov. 3: 1023-1035. de Bodinat C, Guardiola-Lemaitre B, Mocaër E, Renard P, Muñoz C, Millan MJ. 2010. Agomelatine, the first melatonergic antidepressant: discovery, characterization and development. Nat Rev Drug Discov. 9: 628-642. Deacon S, Arendt J. 1996. Adapting to phase shifts, II. Effects of melatonin and conflicting light treatment. Physiol Behav. 59: 675-682. Depaulis A, van Luijtelaar ELJM. 2006. Genetic models of absence epilepsy in the rat. In Pitkanen A, Schwartzkroin PA, Moshe SL (Eds) Models of seizures and epilepsy. Elsevier Academic Press, San Diego, CA, pp. 233–248. Descamps A, Rousset C, Millan MJ, Spedding M, Delagrange P, Cespuglio R. 2009. Influence of the novel antidepressant and melatonin agonist/serotonin2C receptor antagonist, agomelatine, on the rat sleep-wake cycle architecture. Psychopharmacology (Berl). 205: 93-106. 174 Dodd AN, Salathia N, Hall A, Kévei E, Tóth R, Nagy F, Hibberd JM, Millar AJ, Webb AA. 2005. Plant circadian clocks increase photosynthesis, growth, survival, and competitive advantage. Science. 309: 630-633. Drinkenburg WH, Coenen AM, Vossen JM, van Luijtelaar EL. 1991. Spike-wave discharges and sleep-wake states in rats with absence epilepsy. Epilepsy Res 9: 218–224. Duchene D, Vaution C, Glomot F. 1986. Cyclodextrin, their value in pharmaceutical technology. Drug Development & Industrial Pharmacy, 12: 2193-2215. Durazzo TS, Spencer SS, Duckrow RB, Novotny EJ, Spencer DD, Zaveri HP. 2008. Temporal distribution of seizure occurrence from various epileptogenic regions. Neurology 70: 1265–1271. Fisher SP, Davidson K, Kulla A, Sugden D. 2008. Acute sleep-promoting action of the melatonin agonist, ramelteon, in the rat. J Pineal Res. 45: 125-132. Golombek DA, Cardinali DP. 1993. Melatonin accelerates reentrainment after phase advance of the light-dark cycle in Syrian hamsters: antagonism by flumazenil. Chronobiol Int. 10: 435-41. Goodwin GM, Emsley R, Rembry S, Rouillon F, Agomelatine Study Group. 2009. Agomelatine prevents relapse in patients with major depressive disorder without evidence of a discontinuation syndrome: a 24-week randomized, double-blind, placebo-controlled trial. J. Clin. Psychiatry 70: 1128–1137. Grassi-Zucconi G, Semprevivo M, Mocaer E, Kristensson K, Bentivoglio M. 1996. Melatonin and its new agonist S-20098 restore synchronized sleep fragmented by experimental trypanosome infection in the rat. Brain Res Bull. 39: 63-68. Halasz P, Terzano MG, Parrino L. 2002. Spike-wave discharge and the microstructure of sleep-wake continuum in idiopathic generalized epilepsy. Neurophysiol Clin 32: 38–53. Hastings MH, Mead SM, Vindlacheruvu RR, Ebling FJ, Maywood ES, Grosse J. 1992. Non-photic phase shifting of the circadian activity rhythm of Syrian hamsters: the relative potency of arousal and melatonin. Brain Res. 591: 20-26. Hofstra WA, Spetgens WPJ, Leijten FSS, van Rijen PC, Gosselaar P, van der Palen J, de Weerd AW. 2009. Diurnal rhythms in seizures detected by intracranial electrocorticographic monitoring: an observational study. Epilepsy Behav 14: 617–621. Huber R, Deboer T, Schwierin B, Tobler I. 1998. Effect of melatonin on sleep and brain temperature in the Djungarian hamster and the rat. Physiol Behav. 65: 77-82. Illnerová H, Trentini GP, Maslova L. 1989. Melatonin accelerates reentrainment of the circadian rhythm of its own production after an 8-h advance of the light-dark cycle. J Comp Physiol A. 166: 97-102. 175 Knutsson A. 2003. Health disorders of shift workers. Occup Med (Lond). 53: 103-108. Kopp C, Vogel E, Rettori MC, Delagrange P, Guardiola-Lemaitre B, Misslin R. 1998. Effects of a daylight cycle reversal on locomotor activity in several inbred strains of mice. Physiol Behav 63: 577–585. Koster-van Hoffen GC, Mirmiran M, Bos NP, Witting W, Delagrange P, GuardiolaLemaitre B. 1993. Effects of a novel melatonin analog on circadian rhythms of body temperature and activity in young, middle-aged, and old rats. Neurobiol Aging. 14: 565-569. Lancel M, Kerkhof GA. 1989. Effects of repeated sleep deprivation in the dark- or lightperiod on sleep in rats. Physiol Behav. 45:289-297. Langebartels A, Mathias S, Lancel M. 2001. Acute effects of melatonin on spontaneous and picrotoxin-evoked sleep-wake behaviour in the rat. J Sleep Res. 10: 211217. Lapin IP, Mirzaev SM, Ryzov IV, Oxenkrug GF. 1998. Anticonvulsant activity of melatonin against seizures induced by quinolinate, kainate, glutamate, NMDA, and pentylenetetrazole in mice. J Pineal Res. 24: 215-218. Lee ML, Swanson BE, de la Iglesia HO. 2009. Circadian timing of REM sleep is coupled to an oscillator within the dorsomedial suprachiasmatic nucleus. Curr Biol 19: 848–852. Lemmer B, Kern RI, Nold G, Lohrer H. 2002. Jet lag in athletes after eastward and westward time-zone transition. Chronobiol Int. 19: 743-764. Loo H, Hale A, D’haenen H. 2002. Determination of the dose of agomelatine, a melatonergic agonist and selective 5-HT2C antagonist, in the treatment of major depressive disorder: a placebo-controlled dose range study. Int. Clin. Psychopharmacol. 17: 239–247. Martinet L, Guardiola-Lemaitre B, Mocaer E. 1996. Entrainment of circadian rhythms by S-20098, a melatonin agonist, is dose and plasma concentration dependent. Pharmacol Biochem Behav. 54: 713-718. Monnaert V, Tilloy S, Bricout H, Fenart L, Cecchelli R, Monflier E. 2004. Behavior of alpha-, beta-, and gamma-cyclodextrins and their derivatives on an in vitro model of blood-brain barrier. J Pharmacol Exp Ther. 310: 745-751. Mrosovsky N. Masking: history, definitions, and measurement. Chronobiol Int. 16: 415429. Murphy HM, Wideman CH, Nadzam GR. 1998. The role of vasopressin in modulating circadian rhythm responses to phase shifts. Peptides. 19: 1191-1208. Musshoff U, Speckmann EJ. 2003. Diurnal actions of melatonin on epileptic activity in hippocampal slices of rats. Life Sci. 73: 2603-2610. 176 Nagano M, Adachi A, Nakahama K, Nakamura T, Tamada M, Meyer-Bernstein E, Sehgal A, Shigeyoshi Y. 2003. An abrupt shift in the day/night cycle causes desynchrony in the mammalian circadian centre. J Neurosci 23: 6141–6151. Ovchinnikov A, Lüttjohann A, Hramov A, van Luijtelaar G. (2010). An algorithm for real-time detection of spike-wave discharges in rodents. J Neurosci Methods. 194: 172-178. Papp M, Gruca P, Boyer PA, Mocaër E. 2003. Effect of agomelatine in the chronic mild stress model of depression in the rat. Neuropsychopharmacology 28: 694–703. Pavlova MK, Shea SA, Bromfield EB. 2004. Day/night patterns of focal seizures. Epilepsy Behav 5: 44–49. Pittendrigh CS. 1981. Circadian systems: entrainment. In Handbook of Behavioral Neurobiology Vol 4, Biological Rhythms, Eds. Aschoff J. Plenum Press, New York. pp 95-124. Pytel M, Mercik K, Mozrzymas JW. 2006. Interaction between cyclodextrin and neuronal membrane results in modulation of GABA(A) receptor conformational transitions. Br J Pharmacol. 148: 413-422. Quigg M, Straume M, Menaker M, Bertram EH III. 1998. Temporal distribution of partial seizures: comparison of an animal model with human partial epilepsy. Ann Neurol 43: 748–755. Reddy AB, Field MD, Maywood ES, Hastings MH. 2002. Differential resynchronisation of circadian clock gene expression within the suprachiasmatic nuclei of mice subjected to experimental jet lag. J Neurosci 22: 7326–7330. Redman JR, Guardiola-Lemaitre B, Brown M, Delagrange P, Armstrong SM. 1995. Dose dependent effects of S-20098, a melatonin agonist, on direction of reentrainment of rat circadian activity rhythms. Psychopharmacology (Berl). 118: 385-390. Reinberg A, Vieux N, Ghata J, Chaumont AJ, LaPorte A. 1979. Amplitude of the oral temperature circadian rhythm in relation to the ability to phase shift circadian rhythms of shift workers. Chronobiol. 6: 57-63. Sei H, Kiuchi T, Chang HY, Morita Y. 1992. Effects of an eight-hour advance of the lightdark cycle on sleep-wake rhythm in the rat. Neurosci Lett. 137: 161-164. Sei H, Kiuchi T, Chang HY, Seno H, Sano A, Morita Y. 1994. Response of the sleep-wake rhythm to an 8-hour advance of the light-dark cycle in the rat. Chronobiol Int. 11: 293-300. Sharma VK, Singaravel M, Subbaraj R, Chandrashekaran MK. 1999. Timely administration of melatonin accelerates reentrainment to phase-shifted lightdark cycles in the field mouse Mus booduga. Chronobiol Int. 16: 163-170. 177 Shu HJ, Eisenman LN, Jinadasa D, Covey DF, Zorumski CF, Mennerick S. 2004. Slow actions of neuroactive steroids at GABAA receptors. J Neurosci. 24: 6667-6675. Smyk MK, Coenen A, Lewandowski MH, van Luijtelaar G. 2012. desynchronization facilitates seizures. Epilepsia. 53: 1511-1518. Internal Smyk MK, Coenen AM, Lewandowski MH, van Luijtelaar G. 2011. Endogenous rhythm of absence epilepsy: relationship with general motor activity and sleep-wake states. Epilepsy Res. 93: 120-127. Takamure M, Murakami N, Takahashi K, Kuroda H, Etoh T. 1991. Rapid reentrainment of the circadian clock itself, but not the measurable activity rhythms to a new light-dark cycle in the rat. Physiol Behav 50: 443–449. Tobler I, Jaggi K, Borbély AA. 1994. Effects of melatonin and the melatonin receptor agonist S-20098 on the vigilance states, EEG spectra, and cortical temperature in the rat. J Pineal Res. 16: 26-32. Tolmacheva EA, van Luijtelaar G. 2007. Absence seizures are reduced by the enhancement of GABA-ergic inhibition in the hippocampus in WAG/Rij rats. Neurosci Lett. 416: 17-21. Tresguerres JA, Ariznavarreta C, Granados B, Martín M, Villanúa MA, Golombek DA, Cardinali DP. 2001. Circadian urinary 6-sulphatoxymelatonin, cortisol excretion and locomotor activity in airline pilots during transmeridian flights. J Pineal Res. 31: 16-22. van den Heuvel CJ, Ferguson SA, Macchi MM, Dawson D. 2005. Melatonin as a hypnotic: con. Sleep Med Rev. 9: 71-80. van Luijtelaar EL, Coenen AM. 1986. Two types of electrocortical paroxysms in an inbred strain of rats. Neurosci Lett 70:3 93–397. van Luijtelaar ELJM, Coenen AML. 1988. Circadian rhythmicity in absence epilepsy in rats. Epilepsy Res 2: 331–336. van Reeth O, Olivares E, Turek FW, Granjon L, Mocaer E. 1998. Resynchronisation of a diurnal rodent circadian clock accelerated by a melatonin agonist. Neuroreport. 9:1901-1905. van Reeth O, Weibel L, Olivares E, Maccari S, Mocaer E, Turek FW. 2001. Melatonin or a melatonin agonist corrects age-related changes in circadian response to environmental stimulus. Am J Physiol Regul Integr Comp Physiol. 280: 582591. Wang F, Li J, Wu C, Yang J, Xu F, Zhao Q. 2003. The GABA(A) receptor mediates the hypnotic activity of melatonin in rats. Pharmacol Biochem Behav. 74: 573-578. 178 Wang MD, Wahlström G, Bäckström T. 1997. Pregnenolone sulphate and pregnenolone do not interact with 5 beta-pregnanolone- and hexobarbitone-induced anaesthesia in the rat. Br J Anaesth. 78: 328-331. Waterhouse J, Jones K, Edwards B, Harrison Y, Nevill A, Reilly T. 2004. Lack of evidence for a marked endogenous component determining food intake in humans during forced desynchrony. Chronobiol Int. 21: 445-468. Waterhouse J, Reilly T, Atkinson G, Edwards B. 2007. Jet lag: trends and coping strategies. Lancet. 369: 1117-1129. Wegmann HM, Gundel A, Naumann M, Samel A, Schwartz E, Vejvoda M. 1986. Sleep, sleepiness, and circadian rhythmicity in aircrews operating on transatlantic routes. Aviat Space Environ Med. 57: B53-64. Wever R. 1966. The duration of re-entrainment of circadian rhythms after phase shifts of the Zeitgeber. A theoretical investigation. J Theor Biol 13: 187–201. Winget CM, DeRoshia CW, Markley CL, Holley DC. 1984. A review of human physiological and performance changes associated with desynchronosis of biological rhythms. Aviat Space Environ Med. 55: 1085-1096. Yamazaki S, Numano R, Abe M, Hida A, Takahashi R, Ueda M, Block GD, Sakaki Y, Menaker M, Tei H. 2000. Resetting central and peripheral circadian oscillators in transgenic rats. Science 288: 682–685. Yan L. 2011. Structural and functional changes in the suprachiasmatic nucleus following chronic circadian rhythm perturbation. Neuroscience. 183: 99-107. Yildirim M, Marangoz C. 2006. Anticonvulsant effects of melatonin on penicillin-induced epileptiform activity in rats. Brain Res. 1099: 183-188. Zhdanova IV. 2005. Melatonin as a hypnotic: pro. Sleep Med Rev. 9: 51-65. 179 CHAPTER 6 General Discussion Enhancing/inhibiting influences affecting the occurrence of absence seizures may be created internally within the organism (state of vigilance, hormonal environment) or be delivered from the outside (light stimulation, stress, chemical agents) (Wolf and Goosses, 1986; Baykan et al., 2005; Drinkenburg et al., 1991; Lannes et al., 1985; Halász et al., 2002; Grünewald et al., 1992; van Luijtelaar et al., 2001; Tolmacheva et al., 2012; Somerville, 2009). The aim of the present thesis was to establish the contribution of endogenous and exogenous factors to the occurrence of absence seizures. In particular, the endogenous factors that were investigated were the circadian timing system and sleep-wake states, the external contributors were represented by the light-dark cycle and a melatonin agonist. In Chapter 2, the temporal relationship between the commonly distinguished sleep-wake states (active and passive wakefulness, light and deep slow-wave sleep, REM sleep) and the occurrence of SWDs was investigated for the whole 24 h cycle and for the light and the dark phase separately. Endogenous circadian control was examined in Chapter 3, in which WAG/Rij rats were exposed to a constant condition paradigm. Chapter 4 describes consequences of circadian misalignment caused by an abrupt shift in the light-dark cycle. In Chapter 5, first the effects of various doses of acute administration of the melatonin agonist agomelatine on sleep were investigated, next it was attempted to ameliorate the consequences of a similar shift in the light-dark cycle by daily, timed administration of this melatonin agonist. Main findings of the thesis: I. SWDs and sleep-wake states are temporally related. Regardless of the method of data processing (correlations and crosscorrelations across 24 h versus analysis of 5 s preceding the 183 occurrence of SWDs), the relationship between SWDs and deep slow-wave sleep is always negative, while relationship with active wakefulness and light slow-wave sleep varies depending on the approach chosen. II. The character of SWDs – sleep-wake states relationships is determined by the phase of the light-dark cycle. In the light phase, the relationships are unidirectional, which means that the duration of a particular sleep-wake state may serve as a reliable predictor of the duration of SWD activity. In the dark phase however, the relationships are bidirectional. In addition to predictive value of sleep-wake states for this duration, absence seizures themselves influence the duration of sleepwake states. III. SWDs – sleep-wake states relationships estimated on a momentary level (analysis of 5 s EEG preceding and following the occurrence of SWDs) is independent of the state of the circadian timing system: entrained versus fee-running. IV. Rhythmic occurrence of SWDs observed during entrainment to the 12:12 light-dark cycle persists also under constant dim light condition, which strongly suggests that the rhythm is generated endogenously. In the absence of environmental time cues, the rhythm of SWDs free-runs with a period length of about 23 h and desynchronizes from the rhythm of general motor activity. V. Rapid change in the timing of the light phase of the 12:12 lightdark cycle results in circadian desynchronization between rhythms of SWDs, general motor activity and sleep-wake states. Different rhythms re-entrain with different speed, although some rhythms seem coupled. Active wakefulness and deep slow-wave sleep are thought to share a common circadian oscillator since they re-synchronize together. The same is thought for SWDs and light slow-wave sleep. 184 VI. Timed administration of agomelatine, agonist of melatonin MT 1 and MT2 receptors, neither affect the occurrence and duration of SWDs and sleep-wake states during entrainment to the 12:12 light-dark cycle, nor accelerates the speed of re-entrainment of these rhythms after the phase shift of the photoperiod. VII. Alterations in circadian physiology such as internal desynchronization are accompanied by exacerbation of epileptic activity. Changes in the duration of sleep-wake states, deep slow-wave sleep in particular, seems to be responsible for increased number and duration of seizures. Relationship between SWDs and sleep-wake states The first series of questions addressed by the present thesis regarded the relationship between absence seizures and sleep-wake states. It was aimed to establish whether the relationship is constant across 12:12 light-dark cycle and whether it is modulated by the state of the circadian timing system: entrained versus free-running. Vigilance is one of the major factors affecting the incidence of absence seizures in humans and in animal models (Drinkenburg et al., 1991; Lannes et al., 1985; Halász et al., 2002). SWDs in WAG/Rij rats occur mainly during passive wakefulness and light slow-wave sleep, while they are hardly preceded by deep slow-wave sleep and REM sleep (Drinkenburg et al., 1991). In addition, cortical electrical stimulation induces SWD-like afterdischarges mainly during drowsiness, and much less during active wakefulness and slow wave sleep (Lüttjohann and van Luijtelaar, 2012). Relationships described above have been established by inspection of 5 s epochs directly preceding the onset of SWDs (Drinkenburg et al., 1991). This approach has been called “momentary”. 185 In Chapter 2, a link between absence seizures and sleep-wake states was investigated in a different way. Correlations and crosscorrelations between SWDs and all vigilance states were calculated over the 24 h and separately for the light and the dark phase. While the outcomes of the study agreed upon the role of deep slow-wave sleep and, to lesser extent, passive wakefulness and REM sleep, the results where different regarding active wakefulness and light slowwave sleep. Contrary to inhibitory effect revealed by the momentary approach, active wakefulness was positively correlated with the incidence of SWDs. Light slow-wave sleep, found previously to enhance generation of SWDs, showed negative correlation with the occurrence of seizures. A similar discrepancy regarding active wakefulness has been described in inbred Fisher and Brow Norway rats exhibiting generalized neocortical high voltage spike-wave spindles (HVS), an equivalent of SWDs (Jandó et al., 1995). Data analyzed in 10 min bins showed that the peak of HVS incidence was preceded by a peak in activity occurring 10 min earlier. Spectral analysis of delta band in 2 s epochs of EEG directly preceding HVS revealed however, that HVS do not occur during deep slow-wave sleep or a highly aroused state (Jandó et al., 1995). The different methods of data analysis can be considered as the source of this contradiction. Cross-correlations provide information about the global organization of sleep-wake states and the temporal relationships between the different sleep-wake states, while momentary factors provide information about the state just before SWDs. Active wakefulness preceding the occurrence of SWDs in a time range of 25 – 30 min found in the experiment described in the Chapter 2 is not surprising given the fact that one of the most frequent state transitions across rat sleep-wake cycle is passive wakefulness → active wakefulness → passive wakefulness changeover (Gervasoni et al., 2004). SWDs are positively correlated with this particular type of transition as it was shown previously and then confirmed in the study 186 described in Chapter 2. Moreover, the change of active wakefulness into passive wakefulness creates one of the most convenient neuronal environments for the occurrence of SWDs. The sequence: passive wakefulness → slow-wave sleep → intermediate stage → REM sleep → passive wakefulness (Gervasoni et al., 2004) explains also negative cross-correlations between SWDs and both slow-wave sleep states. As mentioned previously, SWDs rarely occur during REM sleep, which usually follows slow-wave sleep. In Chapter 2 differentiation between two slow-wave sleep stages was also made. Both: light → deep slowwave sleep and deep → light slow-wave sleep changeovers were found to be negatively correlated with the incidence of absence seizures across 24 h. Results obtained in the experiment presented in Chapter 2 revealed also that temporal relationships between SWDs and sleepwake states are modified by the phase of the 12:12 light-dark cycle. In the light phase, relationships are stronger and unidirectional, the duration of a particular sleep-wake state predicts the duration of SWDs. In the dark phase however, the presence of SWDs is influenced by, but also influences the duration of sleep-wake states. Clinical and experimental data provide evidence for a reciprocal relationship between sleep and epilepsy (Shouse et al., 2000; Matos et al., 2010). Data collected in a population of patients with temporal lobe epilepsy showed pronounced changes in the amount of sleep states (Bazil et al., 2000). The degree to which the structure and efficiency of sleep was changed was dependent on the time of seizures occurrence (day vs. night). Daytime seizures reduced REM sleep period, while nighttime seizures, besides REM sleep suppression, decreased stage 2 and 4 slow-wave sleep, sleep efficiency, and increased the amount of stage 1 slow-wave sleep. Further reduction of REM sleep was found when seizures occurred before the first REM episode (Bazil et al., 2000). Similar changes have been reported 187 for generalized epilepsy. Polysomnographic studies in a group consisted of childhood absence epilepsy, juvenile absence epilepsy and juvenile myoclonic epilepsy patients revealed a decrease in sleep efficiency and deep slow-wave sleep (stage 4, a human equivalent of deep slow-wave sleep), while wakefulness time, the percentage of stage 1 slow-wave sleep and REM sleep latency were found to be increased (Baretto et al., 2002; Maganti et al., 2005). A time-of-a-day effect has been found in amygdalakindled rats (Yi et al., 2012). Whether homeostatic or circadian mechanisms of the sleep-wake regulation were affected was dependent of the time across the 12:12 light-dark cycle during which the rats were kindled. Although the time distance between kindling and changes in the percentage of sleep states and duration of such changes were different, a common finding was the decrease in both slow-wave and REM sleep (Yi et al., 2012). Common findings for the studies cited above and some of the studies performed on the WAG/Rij model are: the presence of the circadian factor determining the timing of aberrations in the sleep cycle and the reduction of REM sleep. 6-month-old WAG/Rij rats endowed with more SWDs have a shorter sleep cycle in general, and a shorter sleep cycle particularly at the end of the light phase in comparison to 4-month-old strain counterpart, which have significantly less SWDs than older rats (van Luijtelaar and Bikbaev, 2007). It might be speculated that a presence and persistence of a large number of SWDs practically during the whole dark phase may influence sleep variables at some other point of the 24 h cycle. The end of the light phase is the time when light slow-wave sleep predominates. Moreover, as shown by Drinkenburg and co-workers (1991) and also in Chapter 3, SWDs exert an awaking effect since active and passive wakefulness are the most common states following the occurrence of SWDs. Taken together, at the end of the light period when sleep is light and SWDs 188 with awakening properties increase in incidence, the sleep-wake cycle of WAG/Rij rats is rather fragile for interruptions. While temporal relationships between SWDs and sleep-wake states were found to be modified by the phase of the light-dark cycle, momentary factors stayed unchanged regardless of whether the animals were entrained or not. Chapter 3 describes the experiment investigating the potential circadian character of the rhythm of absence seizures. Circadian parameters of the rhythm were assessed in 12:12 light-dark cycle and then compared with those obtained in constant dim light. With regard to well-documented behavior in the constant conditions, rhythm of general motor activity was chosen as a reference for the free-running state of the circadian timing system. Additionally to rhythms parameters, the momentary approach was used to examine SWDs – sleep-wake states relationships. The period length, mean, amplitude and robustness of the free-running SWDs rhythm changed substantially; however, the relationship with sleep-wake states remained the same. The rhythms of absence seizures and general motor activity desynchronized from each other, as indicated by a different period length in constant conditions. Nonetheless, temporal desynchronization had no effect on seizure generation. Passive wakefulness remained the most favorable state for the occurrence of SWDs, whereas active wakefulness and REM sleep were the least encouraging. It might be concluded that while the biological clock governs the timing of the physiological or, as in this case, pathological processes, it is not able to affect the primary mechanisms of the generation of SWDs. Although temporal relationships between different cycles may change as a result of circadian desynchronization, corticothalamo-cortical mechanisms of SWDs generation does not seem to be affected by such manipulations. To illustrate the point: Musshoff and Speckmann (2003) investigated time-of-a-day effect on epileptic activity in hippocampal slices of rats. Epileptiform field potentials (EFP) 189 were elicited by omitting magnesium from the bath solution or by adding bicuculline, a GABAA receptor antagonist, to the bath control solution. Despite a difference in the latency of the occurrence of EFP between slices prepared during the light and the dark phase, an imbalance between inhibitory GABA and excitatory neurotransmission mediated by NMDA receptors was a necessary condition for the generation of EFP (Musshoff and Speckmann, 2003). Similarly in the present study, sleep-wake states classified by the morphology and frequency of brain waves captured by the EEG reflect the state of the cortico-thalamo-cortical circuits. SWDs are generated under conditions of hyperexcitable cortical focus and the thalamus in an oscillatory mode, both criteria were fulfilled more frequently during passive wakefulness, light slow-wave sleep and transitional, unstable periods. Therefore, the timing of sleep-wake states and the temporal relationship between them and absence seizures (both variable under the control of the circadian timing system) may change, however, seizures will not be generated unless the momentary criteria of the particular state of the cortico-thalamo-cortical network are satisfied. Endogenous character of the rhythm of SWDs The second set of questions raised by the present thesis regarded the character of the cyclic occurrence of absence seizures. The possible endogenous origin of the rhythm of SWDs was investigated by means of a constant condition paradigm. The paradigm assumes that endogenous character of the rhythm is revealed after elimination of Zeitgebers. During the protocol, an organism is exposed to either one of the two lightning conditions: constant darkness or constant light, with other periodic environmental influences or periodically occurring factors being abolished (Duffy and Dijk, 2002; Jud et al., 2005). The method is often used in animal research to 190 quantify circadian phenotypes (e.g. to determine parameters of freerunning circadian rhythms) (Ruby et al., 2002; Bittman et al., 2007). In the experiment described in Chapter 3, 12:12 light-dark cycle was replaced by constant dim light. Circadian parameters of the rhythm of SWDs and general motor activity (serving as an indicator of the state of the circadian timing system) were assessed for 10 days before and 10 days after removal of the light-dark cycle. The rhythmic occurrence of SWDs, described previously by van Luijtelaar and Coenen (1988) and confirmed by the present study, persisted also in the absence of this powerful Zeitgeber, however the circadian parameters of the SWDs rhythm changed significantly. The period length decreased to ~ 23 h, which made the rhythm a free-running one, mean and amplitude increased and the robustness decreased. These results showed that the presence of entrainment provided by the light-dark cycle is not necessary for the cyclic expression of SWDs in WAG/Rij rats, therefore it might be speculated that the rhythm is generated endogenously. The presence of light and dark however, enhances this rhythmicity by giving it a well-defined, distinct shape. The constant condition paradigm, precisely, the constant darkness variant, was used in the first study investigating the endogenous character of the rhythm of epileptic seizures in a rat model of human mesial temporal lobe epilepsy (MTLE) (Quigg et al., 2000). The study showed that spontaneous limbic seizures in rats occurred in a 24 h pattern regardless of the presence of the light-dark cycle. Moreover, the phase relationship in the distribution of seizures was also preserved: seizures occurring numerously in the light phase during entrainment were also dominating in the corresponding, passive phase of the constant darkness conditions (Quigg et al., 2000). Outcomes of both studies are very suggestive of the endogenous origin of MTL and absence epilepsy rhythms however; it should be kept in mind that the light-dark cycle is not the only factor 191 capable of entraining or masking. In human circadian research, removal of environmental Zeitgebers is thought to be insufficient in the assessment of the circadian phase. Considering that rhythms are often masked by behavioral states (e.g. temperature rhythms is masked by activity/sleep-wake cycle (Weinert and Waterhouse, 1998; Waterhouse et al., 2000), two methods were developed to remove these influences. Forced desynchronization and constant routine protocols aim at unmasking circadian rhythms by distributing sleep episodes evenly across all circadian phases or by eliminating periodic changes in behavior, respectively. This is done by scheduling subjects to sleepwake cycle that is far outside the range of entrainment (the former), or by means of sleep deprivation, constant posture, identical meals distributed across the circadian cycle additionally to constant external environment (the latter) (Czeisler et al., 1999; Duffy et al., 2002; Hofstra and de Weerd, 2008). A forced desynchronization protocol was used to identify circadian patterns in the occurrence of epileptic seizures in humans (Pavlova et al., 2009). The number of days required to fulfill the demands of the protocol depends on the length of an artificial day which must be outside the range of entrainment. The longer the day (e.g. 28 h), the longer the duration of the protocol to obtain even distribution of sleep-wake states across all circadian phases. The protocol used by the authors was a short day protocol (5 h 40 min) and was completed after 3 days. The study suggested a circadian variation in the occurrence of interictal epileptiform discharges (IEDs), independently of the sleep-wake cycle. It also showed that a strict circadian protocol is feasible and safe in patients with generalized epilepsies (Pavlova et al., 2009). The study described in Chapter 2 did not investigate possible behavioral masking from the side of the sleep-wake states. The relationship between SWDs and 192 vigilance is well established (Drinkenburg et al., 1991) but only by the momentary approach and not from the rhythm point of view. Therefore, a forced desynchrony protocol, e.g. in the form proposed by de la Iglesia and coworkers (2004), would be a valuable complement to the study devoted to circadian nature of the rhythm of absence seizures. Internal desynchronization and epilepsy One of the objectives of the present thesis was to investigate consequences of rapid, change in the 12:12 light-dark cycle, which are known to cause serious disruptions in the circadian timing system. Chapters 3 to 5 describe behavior of absence seizures during internal desynchronization and re-entrainment processes after an 8 h light phase delay. An increase in the number and mean duration of SWDs was observed regardless of the cause of desynchronization (constant dim light or a light phase delay). Based on a well-characterized relationship between absences and sleep-wake states (Drinkenburg et al., 1991), this elevation was attributed to changes in the amount of deep slow-wave sleep (a decrease) and passive wakefulness (an increase). Results of the experiments described in Chapters 4 and 5 (the phase delay paradigm) point to a phase-related redistribution of SWDs so that, after the phase shift, they occurred frequently on an unusual time with respect to their normal 24 h rhythm. The outcomes support the hypothesis that disturbances in sleep-wake cycle are responsible for aggravation of epileptic activity. Signs of circadian disturbances have been identified in a wide range of medical conditions including neurodegenerative disorders (Videnovic et al., 2014), affective disorders (Benca et al., 2009) and cancer (Erren et al., 2008). Moreover, members of 24/7 society challenge their internal clock by irregular work/leisure schedules and frequent jet lag. Recurrent disruptions in the circadian organization 193 entail serious health consequences (Knutsson, 2003). There are large interindividual differences regarding tolerance to internal desynchronization and shift work (Reinberg et al., 2007). Subjects defined as allochronic report neither complaints, nor detectable medical symptoms associated with internal desynchronization, this makes them well-tolerant to non-standard working hours. People defined as dyschronic suffer from circadian desynchronization and therefore, show poor tolerance to shift work (Reinberg et al., 2007; Reinberg and Ashkenazi, 2008). Although a small resistance to internal desynchronization is associated with a small circadian amplitude and a period length differing substantially from 24 h, poor tolerance cannot be assessed a priori (Reinberg and Ashkenazi, 2008). Only exposition to challenging conditions may answer the question about one’s susceptibility. However, evidence collected in a population of shift workers, airline crew members attending transmeridian flights and patients experiencing difficulties after rapid changes of time zones or during the course of shift work, may be helpful in predicting possible health consequences. This knowledge may be also used to create recommendations for those considering commencement of shift work, or for patients that are worried whether a particular work schedule might aggravate seizures, or for distant air travelling that may result in exacerbating symptoms. The number of passengers traveling by air is constantly growing. European Commission reports 842 million people using air transport within borders of 28 EU member countries in 2013 (Eurostat, 2014). At the same time, WHO estimates that 50 million people worldwide suffer from epilepsy, including 6 million European citizens and 300 000 new cases diagnosed each year in this geographical area (Mitchell et al., 2011). People with relatively high seizure frequency (4 seizures per month) have been described as more likely to avoid air travel, nevertheless, an increased number of individuals with active 194 epilepsy traveling by air might be expected (Trevorrow et al., 2006). Seizures are second to dizziness/vertigo among neurological symptoms reported during flights (Sirven et al., 2002). Alonso-Cánovas and coworkers showed that 41 of 77 patients (53.2%) who developed neurological symptoms within the flight or maximum 6 h after landing, experienced seizures (2011), while only 13 passengers from that group had been diagnosed with epilepsy before entering the study. Possible triggers for seizure activity were found in 12 of them and the triggers were missing one or more doses of antiepileptic drugs (AEDs) during days preceding the flight, sleep deprivation and usage of recreational drugs (Alonso-Cánovas et al., 2011). Only one study to date attempted to investigate the effects of air travel on seizure frequency in a population of people with epilepsy (Trevorrow, 2006). The author reported increased number of seizures during post-flight week, with the seizure rate almost doubled and positively correlated with pre-flight values. The direction and distance of flight were also investigated, however no significant differences were found. Although not directly investigated, post-flight sleep disruption has been hypothesized to be a major cause of the increase of seizures frequency found in a group of individuals with epilepsy after air travel (Trevorrow, 2006). Travel recommendations given to passengers dealing with epilepsy and travelling across multiple time zones usually comprise avoiding sleep deprivation, keeping the dosage and timing of medications approximately the same as before the flight, and taking two separate supplies of medications: one for the checked and the other for the hand luggage (Aerospace Medical Association Medical Guidelines Task Force, 2003; www.britishairways.com). Here it is shown that also in case of absence epilepsy precautions regarding sleep should be undertaken or at least considered. Experiments described in the present thesis were performed in naive WAG/Rij rats, with no 195 pharmacological interventions to reduce absence seizures, thus speculating about necessity of maintaining pre-shift drug schedule was not investigated. The issue, possible to be tested experimentally, may be of a particular significance for patients. Besides jet lag, negative consequences of circadian misalignment are a common companion of shift workers. There is no consistency with regard to people with epilepsy performing shift work. Some of the sources advise the patients not to work during the night shift due to possible seizure enhancing effects of sleep deprivation, violation of AEDs schedules or psychosocial factors (LaDou, 1982; Koller, 1996; Costa, 2003). The other suggests that there is no evidence of significant risk for epileptics engaged in non-standard working hours (Nicholson and Auria, 1999). Data regarding evaluation of sickness, accident and work records in people with epilepsy hired by British Steel Corporation compiled by Dasgupta and co-workers (1982) showed that 20 out of 32 workers with epilepsy were eligible for and did shift work (Dasgupta et al., 1982). The 12 who did not were not willing to undertake such a commitment. This was the only significant difference found between workers suffering from epilepsy and healthy controls. The study, however, did not investigate the source of the shift work reluctance in this particular group of employees. Experiments described in the Chapters 3 to 5 describe consequences of internal desynchronization for the course of absence epilepsy in WAG/Rij rats. The results allow some presumptions regarding the occurrence of seizures during circadian disruption, however, none of the studies used a repeated phase shifts paradigm. Forcing epileptic animals to be active during abnormal activity hours or scheduling repeated phase shifts combined with different schedules and dosage of AEDs would enrich the present knowledge of the course of epilepsy during shift work. 196 Melatonin and epilepsy The last but not least issue investigated in the present thesis was a response of absence seizures and sleep-wake states to acute and chronic treatment with the melatonin agonist agomelatine during stable entrainment and after a phase shift. Agomelatine was chosen according to its phase-shifting abilities, modifying influence on sleepwake states in rodents and proven antidepressant activity (Redman et al., 1995; Martinet et al., 1996; Loo et al., 2002; Papp et al., 2003; Descamps et al., 2009; Goodwin 2009). It was assumed that the compound may exert a beneficial effect on absence seizures and the speed of re-synchronization of various sleep-wake and SWDs rhythms by ameliorating sleep disturbances caused by epileptic phenotype of WAG/Rij rats and the phase shift, as well as through reduction of depressive-like symptoms including changes in sleep-wake parameters associated with epileptic activity in this model (van Luijtelaar and Bikbaev, 2007, Sarkisova et al., 2010; Sarkisova and van Luijtelaar, 2011; van Luijtelaar et al., 2012). However neither acute nor chronic treatment with agomelatine appeared effective and none of the variables investigated in the experiment described in Chapter 5: number and duration of SWDs, duration of sleep-wake states and speed of re-entrainment after the phase shift was affected by agomelatine. The ability to regulate sleep and to protect cells from free radicals may be reasons why melatonin might be of interest as a potential AED. The first attempt to use the hormone in patients was undertaken by Anton-Tay in 1971. Melatonin in a dose of 2 mg/day was found to reduce seizure frequency and to normalize EEG of patients (Anton-Tay et al., 1971; Anton-Tay, 1974). Since then many clinical trials as well as basic studies towards potential therapeutic use of melatonin in the treatment of epilepsy were conducted. Results 197 however, were contradictory: melatonin either increased (Sheldon, 1998; Sandyk et al., 1992), had no effect (Ross et al., 2002; Coppola et al., 2004; Gupta et al., 2005; Elkhayat et al., 2010) or reduced the number of seizures (Fauteck et al., 1999; Peled et al., 2001; Uberos et al., 2011; Goldberg-Stern et al., 2012). Regardless of the outcomes, in the majority of studies cited above melatonin treatment improved sleep parameters, which, considering the negative impact of seizures on sleep, appeared beneficial. Studies in humans with epilepsy have obviously some limitations: the small sample size, the open label design (both the researches and participants know which treatment is being administered), a presence of additional neurological deficits, AEDs mono- or polytherapy. Basic investigations upon potential antiepileptic effect of melatonin were conducted mainly on rodent models of epilepsy and seizure models. Similarly to the outcomes of clinical trials, conflicting results were obtained between and within the various models. Melatonin was shown to be effective against seizures induced by a wide range of chemical agents: quinolinic acid, N-methyl-Daspartate (NMDA), pentylenetetrazole glutamate, (PTZ), and kainic penicillin acid, (Lapin et pilocarpine, al., 1998; Bikjdaouene et al., 2003; Yildirim and Marangoz, 2006; Lima et al., 2011; Tchekalarova et al., 2013; Yildirim et al., 2013). An acute, anticonvulsive action of the hormone was described as: an increase of the latency of the first seizure, a decrease of the duration and intensity of the first seizure, attenuation of epileptic activity in general and a decline of the mortality rate (Bikjdaouene et al., 2003; Yildirim and Marangoz, 2006; Yildirim et al., 2013). Chronic melatonin treatment decreased the frequency of seizures occurring spontaneously after status epilepticus induced by kainic acid and pilocarpine (Lima et al., 2011; Tchekalarova et al., 2013). Opposite to these findings, evidence suggesting that melatonin exerts no or even pro-convulsive effect has 198 been found as well. Xu and Stringer (2008) reported no significant changes of the latency to the forelimb clonus, seizure score or mortality rate after melatonin pre-treatment in the PTZ model (Xu and Stringer, 2008). No changes of the seizure threshold were reported by Mares and co-workers in flurothyl model (2013). Aggravation of epileptic activity by melatonin was observed in CA1 pyramidal layer of the hippocampus in vitro (Musshoff and Speckmann, 2003), while blockage of hippocampal MT1b receptors delayed the onset of epileptic activity induced by pilocarpine (Stewart and Leung, 2005). Phase-related differences regarding the pro-epileptic effect of melatonin were found in both studies and diurnal variation in density and sensitivity of melatonin and GABAA receptors were speculated to be responsible for the phase-related effects observed (Musshoff and Speckmann, 2003; Stewart and Leung, 2005). The effects of melatonin agonists in epilepsy were assessed with two compounds: ramelteon and agomelatine. Ramelteon, registered as melatonin-derived drug against insomnia (McGechan and Wellington, 2005), was found to increase the afterdischarge threshold and a decreased afterdischarge duration in hippocampal-kindled rats, as well as decrease the frequency of seizures in Kcna 1-null mice, an epilepsy model showing spontaneous recurrent seizures. However, doses of the compound used in the study: 30 and 100 mg/kg were far above a dose of 8 mg/kg recommended for patients dealing with insomnia (FenoglioSimeone et al., 2009). Agomelatine has been tested in the following models of epilepsy: PTZ, pilocarpine, picrotoxin, strychnine and electroshock model, showing anticonvulsive action only after PTZ and pilocarpine administration. Likewise ramelteon, the effective doses used in the study: 50 and 75 mg/kg, exceeded the recommended dose (25 mg/kg) (Aguiar et al., 2012). The failure of agomelatine in preventing the occurrence of SWDs in the present study might be attributed to relatively low doses of the 199 drug given to animals both during the acute and chronic experiment. As shown above, anticonvulsive effect of melatonin and its analogues was achieved by doses far outside physiological or recommended therapeutic concentration. Agomelatine in 10 mg/kg, the highest dose used in the present study, was however found to provide true entrainment of the activity rhythm and to interfere with the sleep-wake cycle in rats (Martinet et al., 1996; Descamps et al., 2009). It was expected, that the compound would indirectly affect the incidence of absence seizures by changes in the level of vigilance (promote sleep) in the acute, and promote sleep and accelerate the speed of reentrainment in the chronic study. Apart from slightly disadvantageous experimental design of the phase-shift experiment (timed stimulation of melatonin receptors is more effective in phase advance rather than phase delay paradigm), the lack of response to agomelatine might be also a result of inter-strain differences in a density of melatonin receptors and reactivity of melatonin system in the brain of WAG/Rij rats. Significant rat strains differences in the excretion profile of 6sulphatoxymelatonin, a melatonin metabolite, plasma and pineal gland melatonin concentration, and melatonin binding sites in the SCN have been reported (Klante et al., 1999). Also other rodent strains differ in the melatonin content (Goto et al., 1989). On the other hand, a reduction of the amount of circulating melatonin by pinealectomy was found to enhanced epileptogenesis in Wistar rats suggesting, that decreased melatonin level may predispose to seizures (de Lima et al., 2005). Comparative assessment of the melatonin physiology in WAG/Rij rats has the potential to answer the following questions: whether WAG/Rij rats differ in the melatonin content, receptor density, their reactivity and a response to exogenous melatonin receptor antagonists. If so, then it is likely that these differences participate in the development of epileptic phenotype. Experimental phase advance 200 of the light-dark cycle would additionally extend the knowledge of the consequences of circadian disturbances on epileptic activity. Is the rhythm of absence seizures driven by a common or distinct oscillator? The results of the experiments investigating an endogenous character of the rhythm of absence seizures and re-synchronization of sleep-wake states and SWDs to the shifted photoperiod raised the question about a circadian oscillator governing the rhythm of SWDs. Internal desynchronization between the rhythm of absence seizures and general motor activity described in Chapter 3 and confirmed in Chapter 4 and 5, suggests distinct circadian mechanisms engaged in regulation of these processes. Data complemented in Chapter 5 by the analysis of four additional sleep-wake states revealed coupling between active wakefulness and deep slow-wave sleep, and between light slow-wave sleep and SWDs. The outcomes support the presence of distinct oscillators for activity/deep slow-wave sleep and absences, concurrently linking the latter to light slow-wave sleep. Persisting rhythmicity in the occurrence of absence seizures together with substantial changes in circadian parameters of the rhythm in constant dim light, suggests dependency on the light-entrainable oscillator, the SCN. Two well-defined parts of the SCN: ventrolateral and dorsomedial, respectively, were found to govern different sleep-wake states and behavioral rhythms (Cambras et al., 2007). The dorsomedial SCN controls circadian oscillations of REM sleep and core body temperature independently of the light-dark cycle information. Rhythmic occurrence of motor activity and slow-wave sleep are governed by both SCN divisions: dorsomedial and ventrolateral part (Cambras et al. 2007). The latter is the recipient of the pathways from the retina. Considering the marked contribution of the light-dark cycle 201 to the rhythmic occurrence of SWDs presented in Chapter 3 and SWDs - REM disparity in the re-synchronization rate described in Chapter 5, it might be assumed that the rhythm of absence seizures is governed by both oscillators within the SCN. This assumption places the rhythm in a group with active wakefulness and slow-wave sleep. Nonetheless, the present results differentiate between these states and SWDs, and also between both slow-wave sleep stages. Regarding the hierarchical organization of the circadian timing system, it may be speculated that, while common in the SCN, mechanisms governing rhythmical expression or re-synchronization of rhythms investigated, diverge on the level of dependent oscillators. The output pathways by which the master circadian pacemaker regulates the timing of particular sleep-wake states are not fully established. Only the medial preoptic area (MPA), ventrolateral preoptic nuclei (VLPO) and orexin-containing neurons in the posterior hypothalamus were shown to receive direct projections from the SCN (Leak and Moore, 2001; Novak and Nunez, 2000; Chou et al., 2002; Abrahamson et al., 2000). Circadian information to the remaining sleep-wake regulatory centers conveys through the following relay nuclei: the MPA, dorsomedial hypothalamic nuclei (DMH) and subparaventricular zone (SPVZ) (Deurveilher and Semba, 2005). Among structures directly innervated by the SCN, the VLPO seems to be of a particular interest considering the epileptic phenotype of WAG/Rij rats. A role of this hypothalamic nucleus in sleep physiology is well established (Sherin et al., 1996; Szymusiak et al., 1998; Gaus et al., 2002). Region-specificity with regard to two sleep stages was found in a study investigating effects of bilateral, cell-specific lesion of the VLPO on slow-wave and REM sleep (Lu et al., 2000). Loss of sleeppositive neurons in VLPO cell cluster correlated positively with a decrease of the amount of slow-wave sleep and delta power (Lu et al, 2000). Such a parallel was not observed with regard to REM sleep 202 reduction, however when dorsal and medial-extended VLPO were considered, the correlation between the number of cells and slow-wave sleep (and delta) loss became apparent and significant (Lu et al., 2000). Suntsova and co-workers (2007) reported a functional deficiency of the VLPO sleep-promoting system in WAG/Rij rats suggesting that absence seizures may develop as a result of insufficient sleep initiation mechanisms (Suntsova et al., 2007). As mentioned previously, the occurrence of SWDs is strongly and opposed related to the occurrence of both slow-wave sleep stages: deep slow-wave sleep inhibits absences, while light slow-wave sleep enhances their generation (Drinkenburg et al., 1991). Taken this relationship together with the slow-wave sleep-specific character of the VLPO and insufficiency of the sleep-promoting system within these hypothalamic nuclei in WAG/Rij rats, it might be speculated that VLPO is one of the potential candidates to host a dependent oscillator for the rhythm of SWDs. However, it should be noted that while a lesion of the VLPO resulted in a marked decrease in the amount of both slow-wave and REM sleep, the circadian rhythm of the remaining sleep was not affected (Lu et al., 2000). Thus, there might be some other oscillator/oscillators, which impose the rhythmicity on sleep generated by the VLPO. The DMH and SPVZ are both sites with direct projections to the SCN and also sites of efferent projections to the VLPO (Thompson and Swanson, 1998; Leak and Moore, 2001; Deurveilher and Semba, 2005; Nakamura et al., 2008). The structures are involved in a wide range of physiological processes such as feeding, thermoregulation, restactivity cycle, autonomic stress responses, corticosteroids secretion (Kalsbeek et al., 1996; Bali et al., 2005; Saper et al, 2005; Abrahamson and Moore, 2006). A bilateral, neurotoxic lesion of the DMH and SPVZ resulted in a marked reduction of circadian rhythms of slow-wave sleep, REM sleep, motor activity and body temperature (Lu 203 et al., 2001; Chou et al., 2003). The SCN – DMH/SPVZ – VLPO circuitry seems to be a promising option for the search of the circadian oscillator governing the rhythm of absence seizures. Taking into consideration interactions between SWDs and stress hormones, an involvement of the relay nuclei (the DMH, SPVZ) in reactivity to stress and corticosteroid secretion is additionally encouraging. Figure 1. Schematic representation of the neuronal connections between the master circadian pacemaker, the suprachiasmatic nuclei of the hypothalamus (SCN) and the ventolateral peoptic nuclei (VLPO) responsible for sleep initiation and maintenance. An insufficient sleep-promoting mechanism of the VLPO in WAG/Rij rats is linked to the generation of absence seizures (Suntsova et al., 2007). Together with slow-wave sleep/REM sleep regionspecificity of the VLPO and the fact that the rhythm of SWDs is linked to the rhythm of light slow-wave sleep, the VLPO may be considered as a potential oscillator for the rhythm of SWDs. The subparaventricular zone (SPVZ) and dorsomedial hypothalamic nuclei (DMH) serve as relay nuclei. Modified from Paxinos and Watson, 2007 Future directions Basic chronobiological approaches such as constant condition and phase shift paradigms have been used in the present thesis to assess the contribution of the circadian timing system and external factors to the occurrence of absence seizures in WAG/Rij rats. The 204 outcomes of the experiments suggest an endogenous character of the rhythm of SWDs, the rhythm’s dependency on sleep-wake states and photic entrainment; and distinct dependent oscillatory mechanisms governing rhythms of various electroencephalographic states. Considering the functional complexity of the circadian timing system, a multiplicity of factors affecting absence seizures and possible interactions between circadian and epileptic mechanisms, the present results create more research questions. But not only questions, also experimental opportunities to elucidate how the circadian system governs the SWD rhythm. Apart from ideas mentioned in the previous sections such as implementation of the forced desynchrony protocol, a phase advance or various shift work paradigms, and the assessment of the functionality of the melatonin system and the SCN - DMH/SPVZ – VLPO circuitry in WAG/Rij rats, some other, more applicable and patient-oriented studies should be also considered. The rhythmic incidence of absence seizures, with well-defined timing of the maximum and minimum, seems to be a promising target for chronotherapy. The concept is simply to optimize the timing of drug administration to obtain the highest therapeutic effect with minimal or no adverse effects (Reinberg, 1991; Smolensky and Peppas, 2007; Ohdo, 2010). Chronotherapy integrates chronopathology, which deals with a circadian modulation of clinical symptoms of diseases, chronopharmacology, pharmacokinetics and which comprise of pharmacodynamics circadian and aspects of chronotoxicology providing information about cyclic changes in drugs toxicity (Kaur et al., 2013). Chronopharmacological studies towards assessment of pharmacodynamics, pharmacokinetics and toxicity of AEDs with respect to circadian rhythmicity have been undertaken, mostly with valproic acid, an effective AED for various seizure types, including absence seizures (Meinardi et al., 1975; Patel et al., 1982; Klotz and 205 Reimann, 1984; Nakano et al., 1984; Shirkey et al., 1985; Theisohn et al., 1989; Yoshiyama et al., 1989; Ohdo et al., 1991; Ohdo et al., 1992; Reith et al., 2001; Ben-Cherif et al., 2012; Ben-Cherif et al., 2013). Time-of-day differences with regard to absorption, metabolism, distribution, elimination, toxicity and efficacy of AEDs have been found. Attempts to implement the principles of chronopharmacotherapy in treatment of seizures have been undertaken and described in two studies to date. Yegnanarayan and co-workers (2006) showed the efficacy and safety of chronotherapy in patients with tonic-clonic seizures who either did not respond to standard doses of phenytoin or carbamazepine, or showed serum concentrations of these AEDs above a safe and acceptable level (Yegnanarayan et al., 2006). Improved seizure control obtained by an adjustment of the timing of administration of AEDs with a constant total dose of medications in patients with nocturnal and early-morning seizures has been reported (Guilhoto et al., 2011). Both studies support the idea that the finetuned timing of drugs administration with respect to the occurrence of seizures may help to achieve good control of epilepsy and minimize adverse side effects. A pronounced contribution of the circadian timing system to the occurrence of absence seizures in WAG/Rij rats, well-defined relationship between SWDs and sleep-wake states and susceptibility of the rhythm to phase-shifts makes the model a valuable tool in chronopharmacologcial research of anti-absence medications, complex relationships between systems involved in seizures generation, circadian rhythmicity and drugs metabolism; and from a more applicable point of view: an optimal drug administration schedule. References: Abrahamson EE, Leak RK, Moore RY. 2001. The suprachiasmatic nucleus projects to posterior hypothalamic arousal systems. Neuroreport. 12: 435-440. 206 Abrahamson EE, Moore RY. 2006. Lesions of suprachiasmatic nucleus efferents selectively affect rest-activity rhythm. Mol Cell Endocrinol. 252: 46-56. Aerospace Medical Association Medical Guidelines Task Force. 2003. Medical Guidelines for Airline Travel, 2nd ed. Aviat Space Environ Med. 74: A1-19. Aguiar CC, Almeida AB, Araújo PV, Vasconcelos GS, Chaves EM, do Vale OC, Macêdo DS, de Sousa FC, Viana GS, Vasconcelos SM. 2012. Anticonvulsant effects of agomelatine in mice. Epilepsy Behav. 24: 324-328. Alonso-Cánovas A, de Felipe-Mimbrera A, González-Valcárcel J, García-Barragán N, Corral I, Masjuan J. 2011. Neurology at the airport. J Neurol Neurosurg Psychiatry. 82: 981-985. Antón-Tay F, Díaz JL, Fernández-Guardiola A. 1971. On the effect of melatonin upon human brain. Its possible therapeutic implications. Life Sci. 10: 841-850. Antón-Tay F. 1974. Melatonin: effects Psychopharmacol. 11: 315-324. on brain function. Adv Biochem Bali B, Erdélyi F, Szabó G, Kovács KJ. 2005. Visualization of stress-responsive inhibitory circuits in the GAD65-eGFP transgenic mice. Neurosci Lett. 380: 60-65. Barreto JR, Fernandes RM, Sakamoto AC. 2002. Correlation of sleep macrostructure parameters and idiopathic epilepsies. Arq Neuropsiquiatr. 60: 353-357. Baykan B, Matur Z, Gürses C, Aykutlu E, Gökyiğit A. 2005. Typical absence seizures triggered by photosensitivity. Epilepsia. 46:159-163. Bazil CW, Castro LH, Walczak TS. 2000. Reduction of rapid eye movement sleep by diurnal and nocturnal seizures in temporal lobe epilepsy. Arch Neurol. 57: 363368. Benca R, Duncan MJ, Frank E, McClung C, Nelson RJ, Vicentic A. 2009. Biological rhythms, higher brain function, and behavior: Gaps, opportunities, and challenges. Brain Res Rev. 62: 57-70. Ben-Cherif W, Dridi I, Aouam K, Ben-Attia M, Reinberg A, Boughattas NA. 2012. Chronotolerance study of the antiepileptic drug valproic acid in mice. J Circadian Rhythms. 10: 3. Ben-Cherif W, Dridi I, Aouam K, Ben-Attia M, Reinberg A, Boughattas NA. 2013. Circadian variation of Valproic acid pharmacokinetics in mice. Eur J Pharm Sci. 49: 468-473. Bikjdaouene L, Escames G, León J, Ferrer JM, Khaldy H, Vives F, Acuña-Castroviejo D. 2003. Changes in brain amino acids and nitric oxide after melatonin administration in rats with pentylenetetrazole-induced seizures. J Pineal Res. 35: 54-60. 207 Bittman EL, Costello MK, Brewer JM. 2007. Circadian organization of tau mutant hamsters: aftereffects and splitting. J Biol Rhythms. 22: 425-431. Cambras T, Weller JR, Anglès-Pujoràs M, Lee ML, Christopher A, Diez-Noguera A, Krueger JM, da la Iglesia HO. 2007. Circadian desynchronization of core body temperature and sleep stages in the rat. Proc. Natl. Sci. U.S.A. 104: 7634-7639. Chou TC, Bjorkum AA, Gaus SE, Lu J, Scammell TE, Saper CB. 2002. Afferents to the ventrolateral preoptic nucleus. J Neurosci. 22: 977-990. Chou TC, Scammell TE, Gooley JJ, Gaus SE, Saper CB, Lu J. 2003. Critical role of dorsomedial hypothalamic nucleus in a wide range of behavioral circadian rhythms. J Neurosci. 23: 10691-10702. Coppola G, Iervolino G, Mastrosimone M, La Torre G, Ruiu F, Pascotto A. 2004. Melatonin in wake-sleep disorders in children, adolescents and young adults with mental retardation with or without epilepsy: a double-blind, cross-over, placebo-controlled trial. Brain Dev. 26: 373-376. Costa G. 2003. Shift work and occupational medicine: an overview. Occup Med (Lond). 53: 83-88. Czeisler CA, Duffy JF, Shanahan TL, Brown EN, Mitchell JF, Rimmer DW, Ronda JM, Silva EJ, Allan JS, Emens JS, Dijk DJ, Kronauer RE. 1999. Stability, precision, and near-24-hour period of the human circadian pacemaker. Science. 284: 21772181. Dasgupta AK, Saunders M, Dick DJ. 1982. Epilepsy in the British Steel Corporation: an evaluation of sickness, accident, and work records. Br J Ind Med. 39: 145-148. de la Iglesia HO, Cambras T, Schwartz WJ, Díez-Noguera A. 2004. Forced desynchronization of dual circadian oscillators within the rat suprachiasmatic nucleus. Curr Biol. 14: 796-800. de Lima E, Soares JM Jr, del Carmen Sanabria Garrido Y, Gomes Valente S, Priel MR, Chada Baracat E, Abrão Cavalheiro E, da Graça Naffah-Mazzacoratti M, Amado D. 2005. Effects of pinealectomy and the treatment with melatonin on the temporal lobe epilepsy in rats. Brain Res. 1043: 24-31. Descamps A, Rousset C, Millan MJ, Spedding M, Delagrange P, Cespuglio R. 2009. Influence of the novel antidepressant and melatonin agonist/serotonin2C receptor antagonist, agomelatine, on the rat sleep-wake cycle architecture. Psychopharmacology (Berl). 205: 93-106. Deurveilher S, Semba K. 2005. Indirect projections from the suprachiasmatic nucleus to major arousal-promoting cell groups in rat: implications for the circadian control of behavioural state. Neuroscience. 130: 165-183. 208 Drinkenburg WH, Coenen AM, Vossen JM, Van Luijtelaar EL. 1991. Spike-wave discharges and sleep-wake states in rats with absence epilepsy. Epilepsy Res. 9: 218-224. Duffy JF, Dijk DJ. 2002. Getting through to circadian oscillators: why use constant routines? J Biol Rhythms. 17: 4-13. Elkhayat HA, Hassanein SM, Tomoum HY, Abd-Elhamid IA, Asaad T, Elwakkad AS. 2010. Melatonin and sleep-related problems in children with intractable epilepsy. Pediatr Neurol. 42: 249-254. Erren TC, Pape HG, Reiter RJ, Piekarski C. 2008. Chronodisruption and cancer. Naturwissenschaften. 95: 367-382. Fauteck J, Schmidt H, Lerchl A, Kurlemann G, Wittkowski W. 1999. Melatonin in epilepsy: first results of replacement therapy and first clinical results. Biol Signals Recept. 8: 105-110. Fenoglio-Simeone K, Mazarati A, Sefidvash-Hockley S, Shin D, Wilke J, Milligan H, Sankar R, Rho JM, Maganti R. 2009. Anticonvulsant effects of the selective melatonin receptor agonist ramelteon. Epilepsy Behav.16: 52-57. Gaus SE, Strecker RE, Tate BA, Parker RA, Saper CB. 2002. Ventrolateral preoptic nucleus contains sleep-active, galaninergic neurons in multiple mammalian species. Neuroscience. 115: 285-294. Gervasoni D, Lin SC, Ribeiro S, Soares ES, Pantoja J, Nicolelis MA. 2004. Global forebrain dynamics predict rat behavioral states and their transitions. J Neurosci. 24: 11137-11147. Goldberg-Stern H, Oren H, Peled N, Garty BZ. 2012. Effect of melatonin on seizure frequency in intractable epilepsy: a pilot study. J Child Neurol. 27: 1524-1528. Goodwin GM, Emsley R, Rembry S, Rouillon F, Agomelatine Study Group. 2009. Agomelatine prevents relapse in patients with major depressive disorder without evidence of a discontinuation syndrome: a 24-week randomized, double-blind, placebo-controlled trial. J. Clin. Psychiatry 70: 1128–1137. Goto M, Oshima I, Tomita T, Ebihara S. 1989. Melatonin content of the pineal gland in different mouse strains. J Pineal Res. 7: 195-204. Grünewald RA, Aliberti V, Panayiotopoulos CP. 1992. Exacerbation of typical absence seizures by progesterone. Seizure. 1: 137-138. Guilhoto LM, Loddenkemper T, Vendrame M, Bergin A, Bourgeois BF, Kothare SV. 2011. Higher evening antiepileptic drug dose for nocturnal and early-morning seizures. Epilepsy Behav. 20: 334-337. 209 Gupta M, Aneja S, Kohli K. 2005. Add-on melatonin improves sleep behavior in children with epilepsy: randomized, double-blind, placebo-controlled trial. J Child Neurol. 20: 112-115. Halász P, Terzano MG, Parrino L. 2002. Spike-wave discharge and the microstructure of sleep-wake continuum in idiopathic generalised epilepsy. Neurophysiol Clin. 32: 38-53. Hofstra WA, de Weerd AW. 2008. How to assess circadian rhythm in humans: a review of literature. Epilepsy Behav. 13: 438-444. Jandó G, Carpi D, Kandel A, Urioste R, Horvath Z, Pierre E, Vadi D, Vadasz C, Buzsáki G. 1995. Spike-and-wave epilepsy in rats: sex differences and inheritance of physiological traits. Neuroscience. 64: 301-17. Jud C, Schmutz I, Hampp G, Oster H, Albrecht U. 2005. A guideline for analyzing circadian wheel-running behavior in rodents under different lighting conditions. Biol Proced Online. 7:101-116. Kalsbeek A, Drijfhout WJ, Westerink BH, van Heerikhuize JJ, van der Woude TP, van der Vliet J, Buijs RM. 1996. GABA receptors in the region of the dorsomedial hypothalamus of rats are implicated in the control of melatonin and corticosterone release. Neuroendocrinology. 63: 69-78. Kaur G, Phillips C, Wong K, Saini B. 2013. Timing is important in medication administration: a timely review of chronotherapy research. Int J Clin Pharm. 35: 344-358. Klante G, Secci K, Masson-Pévet M, Pévet P, Vivien-Roels B, Steinlechner S, Wollnik F. 1999. Interstrain differences in activity pattern, pineal function, and SCN melatonin receptor density of rats. Am J Physiol. 276: R1078-1086. Klotz U, Reimann IW. 1984. Chronopharmacokinetic study with prolonged infusion of midazolam. Clin Pharmacokinet. 9: 469–474. Knutsson A. 2003. Health disorders of shift workers. Occup Med (Lond). 53: 103-108. Koller M. Occupational health services for shift and night workers. Appl Ergon. 27: 3137. LaDou J, 1982. Health Effects of Shift Work. West J Med. 137: 525–530. Lannes B, Micheletti G, Vergnes M, Marescaux C, Depaulis A, Warter JM. 1988. Relationship between spike-wave discharges and vigilance levels in rats with spontaneous petit mal-like epilepsy. Neurosci Lett. 94: 187-191. Lapin IP, Mirzaev SM, Ryzov IV, Oxenkrug GF. 1998. Anticonvulsant activity of melatonin against seizures induced by quinolinate, kainate, glutamate, NMDA, and pentylenetetrazole in mice. J Pineal Res. 24: 215-218. 210 Leak RK, Moore RY. 2001. Topographic organization of suprachiasmatic nucleus projection neurons. J Comp Neurol. 433: 312-334. Lima E, Cabral FR, Cavalheiro EA, Naffah-Mazzacoratti Mda G, Amado D. 2011. Melatonin administration after pilocarpine-induced status epilepticus: a new way to prevent or attenuate postlesion epilepsy? Epilepsy Behav. 20: 607-612 Loo H, Hale A, D’haenen H. 2002. Determination of the dose of agomelatine, a melatonergic agonist and selective 5-HT2C antagonist, in the treatment of major depressive disorder: a placebo-controlled dose range study. Int. Clin. Psychopharmacol. 17: 239–247. Lu J, Greco MA, Shiromani P, Saper CB. 2000. Effect of lesions of the ventrolateral preoptic nucleus on NREM and REM sleep. J Neurosci. 20: 3830-3842. Lu J, Zhang YH, Chou TC, Gaus SE, Elmquist JK, Shiromani P, Saper CB. 2001. Contrasting effects of ibotenate lesions of the paraventricular nucleus and subparaventricular zone on sleep-wake cycle and temperature regulation. J Neurosci. 21: 4864-4874. Lüttjohann A, van Luijtelaar G. 2012. The dynamics of cortico-thalamo-cortical interactions at the transition from pre-ictal to ictal LFPs in absence epilepsy. Neurobiol Dis. 47: 49-60. Maganti R, Sheth RD, Hermann BP, Weber S, Gidal BE, Fine J. 2005. Sleep architecture in children with idiopathic generalized epilepsy. Epilepsia. 46: 104-109. Mareš J, Stopka P, Nohejlová K, Rokyta R. 2013. Oxidative stress induced by epileptic seizure and its attenuation by melatonin. Physiol Res. 62: S67-74. Martinet L, Guardiola-Lemaitre B, Mocaer E. 1996. Entrainment of circadian rhythms by S-20098, a melatonin agonist, is dose and plasma concentration dependent. Pharmacol Biochem Behav. 54: 713-718. Matos G, Andersen ML, do Valle AC, Tufik S. 2010. The relationship between sleep and epilepsy: evidence from clinical trials and animal models. J Neurol Sci. 295: 1-7. McGechan A, Wellington K. 2005. Ramelteon. CNS Drugs. 19: 1057-1065. Meinardi H, Van Der Kleijn E, Meijer JW, Van Rees H. 1975. Absorption and distribution of antiepileptic drugs. Epilepsia. 16: 353-365. Mitchell G, Deva N, Harkin M, Skinner P, Werthmann A. 2011. Written Declaration pursuant to Rule 123 of the Rules of Procedure on epilepsy. PE464.670v01-00. Musshoff U, Speckmann EJ. 2003. Diurnal actions of melatonin on epileptic activity in hippocampal slices of rats. Life Sci. 73: 2603-2610. 211 Nakamura W, Yamazaki S, Nakamura TJ, Shirakawa T, Block GD, Takumi T. 2008. In vivo monitoring of circadian timing in freely moving mice. Curr Biol. 18: 381385. Nakano S, Watanabe H, Nagai K, Ogawa N. 1984. Circadian stage-dependent changes in diazepam kinetics. Clin Pharmacol Ther. 36: 271–277. Nicholson PJ, D'Auria DA. 1999. Shift work, health, the working time regulations and health assessments. Occup Med (Lond). 49: 127-137. Novak CM, Nunez AA. 2000. A sparse projection from the suprachiasmatic nucleus to the sleep active ventrolateral preoptic area in the rat. Neuroreport. 11: 93-96. Ohdo S, Nakano S, Ogawa N. 1991. Chronopharmacokinetics of valproic acid following constant-rate administration in mice. Chronobiol Int. 8: 35-43. Ohdo S, Nakano S, Ogawa N. 1992. Circadian changes of valproate kinetics depending on meal condition in humans. J Clin Pharmacol. 32: 822–6. Ohdo S. 2010. Chronotherapeutic strategy: Rhythm monitoring, manipulation and disruption. Adv Drug Deliv Rev. 62: 859-875. Papp M, Gruca P, Boyer PA, Mocaër E. 2003. Effect of agomelatine in the chronic mild stress model of depression in the rat. Neuropsychopharmacology 28: 694–703. Patel IH, Venkataramanan R, Levy RH, Viswanathan CT, Ojemann LM. 1982. Diurnal oscillations in plasma protein binding of valproic acid. Epilepsia. 23: 283–90. Pavlova MK, Shea SA, Scheer FA, Bromfield EB. 2009. Is there a circadian variation of epileptiform abnormalities in idiopathic generalized epilepsy? Epilepsy Behav. 16: 461-467. Paxinos G, Watson C. 2007. The rat brain in stereotaxic coordinates, 6th Edition. Elsevier Academic Press. Peled N, Shorer Z, Peled E, Pillar G. 2001. Melatonin effect on seizures in children with severe neurologic deficit disorders. Epilepsia. 42: 1208-1210. Quigg M, Clayburn H, Straume M, Menaker M, Bertram III EH. 2000. Effects of circadian regulation and rest-activity state on spontaneous seizures in rat model of limbic epilepsy. Epilepsia 41: 502-509. Redman J, Armstrong S, Ng KT. 1983. Free-running activity rhythms in the rat: entrainment by melatonin. Science. 219:1089-1091. Reinberg AE. 1991. Concepts of circadian chronopharmacology. Ann N Y Acad Sci. 618: 102-115. Reinberg A, Ashkenazi I. 2008. Internal desynchronization of circadian rhythms and tolerance to shift work. Chronobiol Int. 25: 625-643. 212 Reinberg AE, Ashkenazi I, Smolensky MH. 2007. Euchronism, allochronism, and dyschronism: is internal desynchronization of human circadian rhythms a sign of illness? Chronobiol Int. 24: 553-588. Reith DM, Andrews J, McLaughlin D. 2001. Valproic acid has temporal variability in urinary clearance of metabolites. Chronobiol Int. 18: 123–129. Ross C, Davies P, Whitehouse W. 2002. Melatonin treatment for sleep disorders in children with neurodevelopmental disorders: an observational study. Dev Med Child Neurol. 44: 339-344. Ruby NF, Brennan TJ, Xie X, Cao V, Franken P, Heller HC, O'Hara BF. 2002. Role of melanopsin in circadian responses to light. Science. 298: 2211-2213. Sandyk R, Tsagas N, Anninos PA. 1992Melatonin as a proconvulsive hormone in humans. Int J Neurosci. 63: 125-135. Saper CB, Lu J, Chou TC, Gooley J. 2005. The hypothalamic integrator for circadian rhythms. Trends Neurosci. 28: 152-157. Sarkisova K, van Luijtelaar G. 2011. The WAG/Rij strain: a genetic animal model of absence epilepsy with comorbidity of depression. Prog Neuropsychopharmacol Biol Psychiatry. 35: 854-876. Sarkisova KY, Kuznetsova GD, Kulikov MA, van Luijtelaar G. 2010. Spike-wave discharges are necessary for the expression of behavioral depression-like symptoms. Epilepsia. 51: 146-160. Sheldon SH. 1998. Pro-convulsant effects of oral melatonin in neurologically disabled children. Lancet. 351:1254. Sherin JE, Shiromani PJ, McCarley RW, Saper CB. 1996. Activation of ventrolateral preoptic neurons during sleep. Science. 271: 216-219. Shirkey RJ, Jellett LB, Kappatos DC, Maling TJ, Macdonald A. 1985. Distribution of sodium valproate in normal whole blood and in blood from patients with renal or hepatic disease. Eur J Clin Pharmacol. 28: 447–452. Shouse MN, Farber PR, Staba RJ. 2000. Physiological basis: how NREM sleep components can promote and REM sleep components can suppress seizure discharge propagation. Clin Neurophysiol. 2: S9-S18. Sirven JI, Claypool DW, Sahs KL, Wingerchuk DM, Bortz JJ, Drazkowski J, Caselli R, Zanick D. 2002. Is there a neurologist on this flight? Neurology. 58: 17391744. Somerville ER. 2009. Some treatments cause seizure aggravation in idiopathic epilepsies (especially absence epilepsy). Epilepsia. 8: 31-36. 213 Smolensky MH, Peppas NA. 2007. Chronobiology, drug chronotherapeutics. Adv Drug Deliv Rev. 59: 828-851. delivery, and Stewart LS, Leung LS. 2005. Hippocampal melatonin receptors modulate seizure threshold. Epilepsia. 46: 473-480. Suntsova N, Kumar S, Guzman-Marin R, Alam MN, Szymusiak R, McGinty D. 2009. A role for the preoptic sleep-promoting system in absence epilepsy. Neurobiol Dis. 36: 126-141. Szymusiak R, Alam N, Steininger TL, McGinty D. 1998. Sleep-waking discharge patterns of ventrolateral preoptic/anterior hypothalamic neurons in rats. Brain Res. 803: 178-188. Tchekalarova J, Petkova Z, Pechlivanova D, Moyanova S, Kortenska L, Mitreva R, Lozanov V, Atanasova D, Lazarov N, Stoynev A. 2013. Prophylactic treatment with melatonin after status epilepticus: effects on epileptogenesis, neuronal damage, and behavioral changes in a kainate model of temporal lobe epilepsy. Epilepsy Behav. 27: 174-187. Theisohn M, Assion HJ, Werner U. 1989. Influence of breakfast and day time on the disposition of carbamazepine in healthy volunteers after intake of single dose (600 mg) of a standard or retard preparation of carbamazepine. Chronopharmacology in Therapy of the Epilepsies. New York: Raven Press; pp. 31–54. Thompson RH, Swanson LW. 1998. Organization of inputs to the dorsomedial nucleus of the hypothalamus: a reexamination with Fluorogold and PHAL in the rat. Brain Res Brain Res Rev. 27: 89-118. Tolmacheva EA, Oitzl MS, van Luijtelaar G. 2012. Stress, glucocorticoids and absences in a genetic epilepsy model. Horm Behav. 61: 706-710. Trevorrow T. 2006. Air travel and seizure frequency for individuals with epilepsy. Seizure. 15: 320-327. Uberos J, Augustin-Morales MC, Molina Carballo A, Florido J, Narbona E, Muñoz-Hoyos A. 2011. Normalization of the sleep-wake pattern and melatonin and 6sulphatoxy-melatonin levels after a therapeutic trial with melatonin in children with severe epilepsy. J Pineal Res. 50: 192-196. Van Luijtelaar EL, Coenen AM. 1988. Circadian rhythmicity in absence epilepsy in rats. Epilepsy Res. 2: 331-336. van Luijtelaar G, Budziszewska B, Jaworska-Feil L, Ellis J, Coenen A, Lasoń W. 2001. The ovarian hormones and absence epilepsy: a long-term EEG study and pharmacological effects in a genetic absence epilepsy model. Epilepsy Res. 46: 225-239. 214 van Luijtelaar G, Bikbaev A. 2007. Midfrequency cortico-thalamic oscillations and the sleep cycle: genetic, time of day and age effects. Epilepsy Res. 73: 259-265. van Luijtelaar G, Wilde M, Citraro R, Scicchitano F, van Rijn C. 2012. Does antiepileptogenesis affect sleep in genetic epileptic rats? Int J Psychophysiol. 85: 49-54. Videnovic A, Lazar AS, Barker RA, Overeem S. 2014. 'The clocks that time us'--circadian rhythms in neurodegenerative disorders. Nat Rev Neurol. 10: 683-693. Waterhouse J, Weinert D, Minors D, Folkard S, Owens D, Atkinson G, Macdonald I, Sytnik N, Tucker P, Reilly T. 2000. A comparison of some different methods for purifying core temperature data from humans. Chronobiol. Int. 17:539–566 Weinert D, Waterhouse J. 1998. Diurnally changing effects of locomotor activity on body temperature in laboratory mice. Physiol Behav. 63:837-43. Wolf P, Goosses R. 1986. Relation of photosensitivity to epileptic syndromes. J Neurol Neurosurg Psychiatry. 49: 1386–1391. Xu K, Stringer JL. 2008. Antioxidants and free radical scavengers do not consistently delay seizure onset in animal models of acute seizures. Epilepsy Behav. 13: 7782. Yegnanarayan R, Mahesh SD, Sangle S. 2006. Chronotherapeutic dose schedule of phenytoin and carbamazepine in epileptic patients. Chronobiol Int. 23: 10351046. Yi PL, Chen YJ, Lin CT, Chang FC. 2012. Occurrence of epilepsy at different zeitgeber times alters sleep homeostasis differently in rats. Sleep. 35: 1651-1665. Yildirim M, Aydin-Abidin S, Abidin I, Akca M, Canpolat S, Cansu A. 2013. Evaluation of the role of chronic daily melatonin administration and pinealectomy on penicillin-induced focal epileptiform activity and spectral analysis of ECoG in rats: an in vivo electrophysiological study. Neurochem Res. 38: 1672-1685. Yildirim M, Marangoz C. 2006. Anticonvulsant effects of melatonin on penicillin-induced epileptiform activity in rats. Brain Res. 1099: 183-188. Yoshiyama Y, Nakano S, Ogawa N. 1989. Chronopharmacokinetic study of valproic acid in man: comparison of oral and rectal administration. J Clin Pharmacol. 29: 1048–1052. 215 ADDENDUM English summary Rhythm in the occurrence of absence seizures in WAG/Rij rats, its origin and interactions with different factors comprise the topic of the present thesis. The experiments described were aimed to answer the question whether the rhythm of SWDs is generated endogenously by the circadian timing system and how the rhythm synchronizes to the most powerful circadian Zeitgeber, the light-dark cycle. Other questions regarded the temporal relationship between SWDs and sleep-wake states, and to which of the sleep-wake rhythms absences are coupled to; finally, whether the melatonin agonist agomelatine is able to influence both SWDs themselves and their rhythm. The thesis provides an insight into circadian aspects of the occurrence of absence seizures, it allows also some predictions about behavior and frequency of absences during regular conditions and during a circadian challenge such as internal desynchronization. Chapter 1 provides general information from the field of chronobiology including time ranges of physiological processes and overview of the mammalian circadian timing system with an emphasis on the pineal hormone melatonin. Examples of 24-h variation in symptoms of common diseases are presented. Next, a general introduction to epilepsy with a particular focus on childhood absence epilepsy and non-random patterns of seizures occurrence is given. The Chapter provides a brief description of various rodent models of childhood absence epilepsy concentrating subsequently on the WAG/Rij strain of rats. The experiments were performed in rats of this strain: the WAG/Rij model is considered as one of the best characterized and validated models. The aims and outline of the thesis are given at the end of the Chapter. In Chapter 2, different, phase-related temporal relationships between SWDs and five sleep-wake states (active wakefulness, passive 219 wakefulness, light and deep slow-wave sleep, REM sleep) were described. The course of epileptic activity and vigilance states was investigated across 24 h, as well as separately for the light and the dark phase of the 12:12 light-dark cycle. Temporal relationships between the five EEG behavioral states and SWDs were assessed by means of correlation and cross-correlation analyses. Differences with regard to the data resolution and the phase of the photoperiod were found. SWDs correlated negatively with deep slow-wave sleep and positively with active wakefulness. An increase in the resolution of the data from 1h to 0.5 min was accompanied by a decrease in the strength of the correlations; this was interpreted as the result of decreased temporal relationship between variables. The character of the relationships appeared to be phase-dependent: unidirectional in the light phase and bidirectional in the dark phase, which suggests different regulatory mechanisms governing the temporal organization of sleep, wakefulness and absence seizures. The endogenous character of the rhythm of SWDs is demonstrated in Chapter 3. WAG/Rij rats, kept initially in 12:12 lightdark cycle, were deprived of this potent circadian Zeitgeber by changing the light regime into constant dim light (20 days). The presence of circadian rhythms in the occurrence of SWDs and general motor activity in both experimental conditions was assessed by the Cosinor method. Additionally, sleep-wake states preceding and following SWDs were identified to establish whether the well-known relationship between SWDs and sleep-wake states is influenced by the circadian timing system. Both rhythms were detected in the constant dim light conditions, however, their parameters changed substantially. Although still present, the rhythm of SWDs lost some of its organization which emphasized the role of the photic entrainment in the timed occurrence of SWDs. Internal desynchronization between SWDs and general motor activity was found suggesting that the rhythms are 220 controlled by relationship different, between dependent SWDs and circadian sleep-wake mechanism. states The established previously was not affected in the constant dim light condition. The matter of internal desynchronization and dependent oscillators governing rhythms of absence seizures and sleep-wake states is further investigated in Chapters 4 and 5. Chapter 4 describes the process of resynchronization of the rhythms of SWDs and general motor activity in WAG/Rij rats subjected to an 8 h light phase delay. Differences regarding the speed and character of re-entrainment were found. After immediate advance, the acrophase of the general motor activity rhythm gradually reached its pre-shift position on the seventh post shift day, whereas the resynchronization of the rhythm of SWDs lasted one day longer and was discontinuous. Internal desynchronization between the rhythms was accompanied by an increase in the number and duration of SWDs; this increase occurred exclusively in the new light phase. To get a detailed insight into the resynchronization process of the rhythm of SWDs and to identify factors responsible for aggravation of epileptic activity, analysis of four remaining sleep-wake states was added to the study described in Chapter 5. Additionally, an attempt to alleviate negative consequences of internal desynchronization was undertaken by timed administration of a melatonin agonist agomelatine. The procedure of the phase shift was identical as presented in Chapter 4. WAG/Rij rats were subjected to the 8 h light phase delay, after which the resynchronization process of the whole sleep-wake cycle was investigated for 10 days. Different speed of reentrainment and coupling between various rhythms was found. The rhythm of SWDs resynchronized together with the rhythm of light slowwave sleep, whereas they uncoupled from the active wakefulness deep slow-wave sleep pair. Again, an increase in the number and duration of SWDs was noted, explained by post shift changes in the 221 amount of passive wakefulness and deep slow-wave sleep. As previously established and confirmed in the chapters of this thesis, passive wakefulness, contrary to deep slow-wave sleep, is the most favorable state of vigilance for the occurrence of SWDs. The melatonin agonist agomelatine had no effects on sleep-wake parameter. Next, the timed administration of agomelatine failed to improve the speed of re-synchronization, it showed also no effect on SWDs during condition of entrainment to the 12:12 light-dark cycle. The thesis ends with Chapter 6, in which all experimental results are evaluated and critically discussed in the view of the main questions addressed in this thesis. It is concluded that the rhythm of SWDs is generated endogenously by the circadian timing system rather than it is imposed by the presence of the light-dark cycle. However, this potent Zeitgeber, has a great impact on the organization of the SWD rhythm. Disturbances in the photic entrainment, either chronic (the constant dim light condition) or acute (phase shift) result in internal desynchronization between the rhythm of SWDs and sleep-wake states, which in turn lead to an increased epileptic activity. The presence of a stable light-dark cycle is an important factor contributing to the occurrence of SWDs, so does the momentary state of vigilance. Apart from momentary relationships, SWDs and sleep-wake states are temporally related. This new finding implies that the duration of particular sleep-wake states can be used as a predictor of the duration of SWDs. While the momentary relationships are constant across the conditions of entrainment and during free-running state of the circadian timing system, temporal relationships between sleep-wake stages inclusive SWDs during entrainment are modulated by the phase of the light-dark cycle. SWDs are particularly related to both slow-wave sleep stages, which is especially apparent during the resynchronization process after the phase shift in the 12:12 light-dark cycle. The rhythm of SWDs re222 entrains together with light slow-wave sleep, while marked increase in the number and duration of absence seizures observed after the phase shift may be attributed to a reduction in the amount of deep slow-wave sleep. Another factor investigated in the thesis, a melatonin agonist agomelatine was found ineffective in influencing SWDs, both in the acute and chronic study, as well as directly and indirectly through sleep-wake states or through interactions with the serotonin system. Results of the experiments described in the thesis generate a number of research questions regarding chronobiological aspects of the occurrence of epileptic seizures. Potential directions of the future studies are presented at the end of Chapter 6. 223 Nederlandse samenvatting Ritmiek in de aanwezigheid, de oorsprong en omgevingsinteracties van absence seizures (aanvallen), zoals die bij WAG/Rij ratten voorkomen, is het onderwerp van dit proefschrift. De beschreven experimenten hebben als doel om de vraag te beantwoorden of het ritme van de piekgolf ontladingen (PGO’s), typerend voor deze aanvallen endogeen opgewekt wordt en hoe het ritme gesynchroniseerd wordt aan de meest krachtige circadiane Zeitgeber, de licht donker cyclus. Andere vragen betreft de temporele relatie tussen PGO’s en de slaap-waak stadia, en aan welk van de slaap-waak ritmes het ritme van PGO’s gekoppeld is; tenslotte, of de melatonine agonist agomelatine een effect heeft op de aanwezigheid van PGO’s zelf, op het ritme van PGO’s en op de slaap-waak stadia. Dit proefschrift geeft inzicht in de circadiane aspecten in het optreden van de aanvallen, het geeft een paar voorspellingen over eigenschappen en de frequentie van de aanvallen tijdens normale omstandigheden en tijdens een aangebrachte verstoring zoals het geval is bij interne desynchronisatie. Hoofdstuk 1 geeft algemene informatie uit het domein van de chronobiologie inclusief de tijdvensters waarin fysiologische processen zich herhalen en een overzicht van het circadiane tijd systeem van zoogdieren met een nadruk of het pijnappelklier hormoon melatonine. Voorbeelden van 24 uur variatie in symptomen van veel voorkomende ziektes worden gegeven. Vervolgens wordt het onderwerp epilepsie ingeleid, met de nadruk op absence epilepsie, zoals dat bij kinderen voorkomt (childhood absence epilepsy, CAE) en op de niet-willekeurige aanwezigheid van de aanvallen bij deze kinderen. Het hoofdstuk geeft verder een korte beschrijving van de verschillende knaagdier modellen van CAE waarbij vervolgens de nadruk ligt op het WAG/Rij ratten model. De experimenten zijn uitgevoerd met dit ratten van deze stam: 224 het WAG/Rij model is een van de best getypeerde en gevalideerde modellen. De vraagstellingen en de verdere opzet van het proefschrift worden aan het eind van het hoofdstuk gepresenteerd. In hoofdstuk 2 werden verschillende, fase gerelateerde temporele relaties tussen absences enerzijds en de vijf slaap-waak stadia (actieve waakzaamheid, passieve waakzaamheid, lichte en diepe („slow wave”) slaap en REM-slaap) anderzijds beschreven. Het verloop van de epileptische activiteit en de vijf stadia werd onderzocht over 24 uur, alsmede afzonderlijk voor de licht en de donker fase van de ieder durende 12:12 licht-donker cyclus. Temporele relaties tussen de verschillende EEG-gedrag gedefinieerde stadia en de PGO’s werden bepaald met behulp van correlatie en kruis-correlatie analyses. Er werden verschillen gevonden betreft de gegevensresolutie en de fase van de fotoperiode. Negatief gecorreleerd met PGO’s is diepe slowwave slaap terwijl actief waakzaamheid en PGO’s positief gecorreleerd zijn. Een verhoging van de resolutie van de gegevens van 1 uur tot 0,5 min ging gepaard met een daling van de sterkte van de correlaties; dit werd geïnterpreteerd als het resultaat van verminderde temporele relatie tussen de stadia. Het karakter van de relaties bleek faseafhankelijk te zijn: unidirectioneel in licht periode en bidirectioneel in het donker. Dit suggereert dat er in deze twee periodes verschillende regulerende mechanismen zijn voor de temporele organisatie van slaap, wakker zijn en PGO’s. Het endogene karakter van het ritme van PGO’s wordt in Hoofdstuk 3 aangetoond. WAG/Rij ratten, die aanvankelijk in een 24h 12:12 licht-donker cyclus gehuisvest werden, werden van hun krachtige circadiane Zeitgeber beroofd door hen gedurende 20 dagen in constant gedimd licht te houden. De aanwezigheid van circadiane ritmes van PGO’s en van algehele motorische activiteit in zowel de eerste als in de tweede periode werd bepaald met behulp van de Cosinor methode. Ook werden de slaap-wake stadia voorafgaand en 225 na afloop van de PGO’s bepaald teneinde vast te stellen of de bekende relatie tussen PGO’s en de vijf slaap-waak stadia beinvloed werd door het circadiaanse tijd sysyeem. Beide ritmes werden gedetecteerd tijdens de standaard huisvesting, de 12-12 periode, als tijdens de constante schemerige lichtomstandigheden. Echter, de parameters van de ritmes waren wezenlijk verschillend. Hoewel nog steeds aanwezig, het ritme van PGO’s verloor wat van zijn strakke temporele organisatie en dit benadrukt de rol van licht in het beinvloeden van het door het endogene ritme opgelegde en daardoor getimde optreden van de PGO’s. Interne desynchronisatie tussen PGO’s en algehele motorische activiteit werd gevonden; dit suggereert dat de ritmes worden gecontroleerd door verschillende, afhankelijk circadiane mechanismes. De relatie tussen PGO’s en de vijf slaap-waak stadia, zoals die eerder was vastgesteld, werd niet differentieel beïnvloed in de constante schemerige lichtomstandigheden. De kwestie van de interne desynchronisatie en afhankelijkheid van de oscillatoren die verantwoordelijk zijn voor de ritmes van de aanvallen en slaap-waak toestanden, werd verder onderzocht in de hoofdstukken 4 en 5. Hoofdstuk 4 beschrijft het proces van resynchronisatie van de ritmes van PGO’s en algehele motorische activiteit in WAG/Rij ratten die blootgesteld werden aan een 8 uur fase verlenging van de licht periode (phase vertraging). Er werden verschillen in snelheid en aard van de resynchronisatie gevonden. De acrophase van het algehele motorische activiteit ritme verschoof geleidelijk en bereikte zijn pre-shift positie op de zevende dag na de verschuiving. De resynchronisatie van het ritme van PGO’s duurde een dag langer en was discontinu. Interne desynchronisatie tussen de ritmes werd vergezeld door een toename van het aantal en de duur van de PGO’s, deze toename vond exclusief plaats in de nieuwe licht periode. m tot een gedetailleerd inzicht in het resynchronisatie proces van het ritme van PGO’s te kunnen komen en om factoren, die 226 verantwoordelijk zijn voor de toename van het aantal aanvallen te kunnen identificeren, werd het experiment bij een nieuwe groep dieren herhaald en zijn de ook de niet eerder geanalyseerde slaap-waak stadia meegenomen en geanalyseerd, de studie is beschreven in Hoofsdtuk 5. Daarnaast is onderzocht of de negatieve gevolgen van interne desynchronisatie verlicht kunnen worden door de herhaalde en getimede toediening van de melatonine agonist agomelatine. De procedure van de faseverschuiving was identiek aan die welke gepresenteerd is in hoofdstuk 4. Naïeve WAG/Rij ratten werden wederom onderworpen aan de 8 uur fase vertraging en het resynchronisatie proces van de hele slaap-waak cyclus werd gedurende 10 dagen onderzocht. Verschillende snelheid van re-synchronizatie en koppelingen tussen verschillende ritmes werden gevonden. Het ritme van PGO’s was gesynchroniseerd aan het ritme van lichte „slow-wave” slaap, terwijl het PGO ritme juist losgekoppeld was van het gepaarde ritme van actief waakzaakheid en diepe „slow-wave” slaap. Er werd wederom een toename van het aantal en de duur van de PGO’s opgemerkt: dit kon worden toegeschreven worden aan de toename in de hoeveelheid passief waakzaamheid en de afname in diepe „slowwave” slaap. Zoals eerder vastgesteld en bevestigd in de hoofdstukken van dit proefschrift, passief waakzaamheid is, in tegenstelling tot diepe „slow-wave” slaap, de toetand die het meest gunstig is voor het optreden van PGO’s. De melatonine agonist agomelatine had noch een effect op de slaap-waak parameters, noch op de snelheid van het resynchronisatie proces, noch op het aantal PGO’s tijdens de 10 daagse meting volgend op de fase verschuiving. Het proefschrift eindigt met Hoofdstuk 6, hier worden alle experimentele bevindingen geevalueerd en tegen het licht gehouden tegen de achtergrond van de belangrijkste vragen zoals die in de inleiding geformuleerd zijn. Geconcludeerd wordt dat het ritme van PGO’s endogeen bepaald wordt en niet opgelegd wordt door de 227 afwisseling van licht en donker. Deze krachtige Zeitgever heeft echter veel impact op de organisatie van het ritme: verstoringen in de vorm van de afwezigheid van deze Zeitgeber, zowel chronisch (tijdens de constante schemer conditie) of acuut (in de twee fase verschuivings experimenten) hadden altijd interne desynchronizatie tussen de ritmes van PGO’s en de slaap stadia tot gevolg, dat op zijn beurt weer aanleiding gaf tot een toename van het aantal aanvallen. De constante aanwezigheid van de licht-donker cyclus is daarom een belangrijke factor die bijdraagt aan de aanwezigheid van PGOs, alsmede het momentane slaap-waak stadium. De slaap-waak stadia en PGO’s zijn op twee manieren met elkaar verbonden: naast de momentane preferentie van PGO’s op vooral tijdens passief waakzaamheid voor te komen, is een nieuwe bevinding dat de duur van de verschillende slaap-waak stadia gebruikt worden als een voorspeller van de duur van PGOs. Terwijl de momentane relaties tussen PGO’s en de slaap stadia altijd hetzelfde zijn, ongeacht of het in base-line condities of constant schemer betreft, de temporele relaties tussen de slaapstadia onderling inclusief die met PGO’s gedurende entrainment worden gemoduleerd afhankelijk van of het licht of donker is. PGO’s zijn met enige nadruk gekoppeld aan zowel lichte and diepe „slow-wave” slaap, hetgeen vooral nog eens extra benadrukt wordt tijdens het resynchronisatie proces na de fase vertraging in de 12:12 licht-donker cyclus. Het ritme van PGO’s loopt tijdens dit resynchronisatie proces in de pas met het ritme van lichte „slow wave” slaap, terwijl de pregnante toename in het aantal PGO’s na de fase verschuiving toegeschreven kan worden aan de afname in de diepe „slow-wave” slaap. Een andere vraag die in het proefschrift onderzocht is, is of de melatonine agonist agomelatine effectief is in het beinvloeden van slaap, PGO’s en de aanpassing na verstoringen. Zowel in de acute als in de chronische studie werden geen directe of indirecte 228 effecten vastgesteld op slaap-waak parameters, of via interakties met het serotonine systeem. Tenslotte, de resultaten van de experimenten beschreven in het proefschrift genereren chronobiologische een aspecten aantal van het onderzoeksvragen optreden van over epileptische aanvallen. Mogelijke richtingen van toekomstig onderzoek worden op het einde van Hoofdstuk 6 gepresenteerd. 229 Curriculum vitae Magdalena Kinga Smyk was born on 7th of October 1983 in Rzeszów, Poland. There, in 2002, she completed the Secondary School of General Education. In the same year she started studies at the Jagiellonian University in Krakow, Faculty of Biology and the Earth Sciences. In 2007 she obtained the MSc degree in Biology, majoring in Neurobiology, with the thesis entitled “Effects of brain injury on status epilepticus induced in rats with no susceptibility to spontaneous epileptic seizures”, supervised by dr. Zuzanna Setkowicz. She started her doctoral studies in 2007 at the Faculty of Biology and the Earth Sciences of Jagiellonian University in Krakow, in the Department of Neurophysiology and Chronobiology, under the supervision of Prof. dr. Marian H. Lewandowski. One year later she became a fellow PhD student in the Donders Centre for Cognition, Radboud University Nijmegen. Under the supervision of Prof. dr. Gilles van Luijtelaar, Prof. dr. Anton Coenen, and Prof. dr. Marian Lewandowski she worked on the PhD project investigating the relationship between absence epilepsy and the circadian timing system finalized and presented in the present thesis. Schools and courses 2014 12th International Course on Epilepsy: Advance International Course: Bridging Basic with Clinical Epileptology. 20 July – 1 August, San Servolo, Venice, Italy 2013 14th International Course on Chronopharmacology. 29 July – 2 August, Mannheim, Germany 2012 IBRO–VLTP Summer School. 27 June–5 July, Krakow, Poland 230 Awards 2015 Travel grant 9th IBRO World Congress of Neuroscience, Rio de Janeiro, Brazil 2014 Travel grant 12th International Course on Epilepsy: Advanced International Course: Bridging Basic with Clinical Epileptology, San Servolo, Venice, Italy 2013 Young Investigator Award, 11 th International Congress of the Polish Neuroscience Society 2013 Travel grant Understanding the of diurnality, Strasbourg, France 2013 Travel grant Society – Environment – Technology, Project of the Jagiellonian University for attending 14 th International Summer Course on Chronopharmacology, Mannheim, Germany 2011 Travel grant, 8th IBRO World Congress of Neuroscience, Florence, Italy 2011 Travel grant 3rd World Congress of Chronobiology, Puebla de Zaragoza, Mexico 2011 Best Student Presentation Prize, Neuroscience Forum, Kraków, Poland neuronal IBRO basis Young List of publications International peer reviewed journals Smyk MK, Coenen A, Lewandowski MH, van Luijtelaar G. 2012. Internal desynchronization facilitates seizures. Epilepsia. 53: 1511-1518. Smyk MK, Coenen A, Lewandowski MH, van Luijtelaar G. 2011. Endogenous rhythm of absence epilepsy: Relationship with 231 general motor activity Research. 93: 120-127. and sleep-wake states. Epilepsy Book chapters Smyk MK, van Luijtelaar G, Drinkenburg WH. 2014. Differential ultradian organization of vigilance states in the light and the dark phase. In: Sleep-wake research in the Netherlands. Annual proceedings of the NSWO. Eds. Tom de Boer, Viviane van Kasteel, Cathalijn Leenaars, Peter Meerlo, Els Most, Floor van Oosterhout and Johan Verbraecken. 25: 77-80. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2010. Spike-wave discharges and sleep-wake states in entrained and free-running conditions. In: Sleep-wake research in the Netherlands. Annual proceedings of the NSWO. Eds. Tom de Boer, Viviane van Kasteel, Birgit Koch, Gilles van Luijtelaar and Johan Verbraecken. 21: 94-97. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2010. Endogenny rytm wyładowań epileptycznych mózgowia szczura w kontekście rytmu ogólnej aktywności motorycznej. In: Techniki elektrofizjologiczne w badaniach zjawisk bioelektrycznych: od kanałów jonowych po sieci neuronalne. Ed. Maria Stankiewicz. Wydawnictwo naukowe Uniwersytetu Mikołaja Kopernika. Toruń. 119-131. Conferences contributions Oral presentations Smyk MK, van Luijtelaar G, Drinkenburg WH. 2013. Old rhythm new methodology: differential organization of sleep-wake states in the light and the dark phase. 11th International Congress of the Polish Neuroscience Society. 15 – 17 September 2013. Poznań, Poland. 232 Smyk MK. 2013. Together but apart: relationship between absence seizures and motor activity. Neuronus IBRO & IRUN Neuroscience Forum. 9 – 11 May 2013. Kraków, Poland. Smyk MK. 2011. Epileptic clock: Circadian control of absence seizures. IBRO Young Neuroscience Forum Neuronus. 15 – 17 April 2011. Kraków, Poland. Smyk MK. 2011. Shifting the clock: seizure response to phase delay. Krakow Workshop on Psychophysiology. 25 – 27 May 2011. Krakow, Poland. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2010. Endogenny rytm wyładowań epileptycznych mózgowia szczura w kontekście rytmu ogólnej aktywności motorycznej. VII Konferencja – Techniki elektrofizjologiczne w badaniach zjawisk bioelektrycznych: od kanałów jonowych po sieci neuronalne. 4 – 5 czerwca 2010. Toruń, Polska. Smyk MK. 2009. Circadian rhythmicity and absence epilepsy. Krakow Workshop on Psychophysiology. 17 – 19 June 2009. Krakow, Poland. Smyk MK. 2008. Future experiments on circadian rhythmicity and absence epilepsy. Krakow Workshop on Psychophysiology. 15 – 18 April 2008. Krakow, Poland. Poster presentations van Luijtelaar G, Smyk MK, Huysmans Seizures, sleep-wake states and antidepressant help to restore International Epilepsy Congress, Istanbul, Turkey. H, Drinkenburg WH. 2015. jet lag: Does melatonergic internal synchrony? 31st 5 – 9 September 2015. Smyk MK, van Luijtelaar G, Huysmans H, Drinkenburg WH. 2015. Agomelatine does not improve speed of re-entrainment of rat sleep-wake states and absence seizures after a phase delay. 9th IBRO World Congress of Neuroscience. 7 – 11 July 2015. Rio de Janeiro, Brazil. 233 Smyk MK, van Luijtelaar G, Drinkenburg WH. 2015. Sleep-wake states across 24 h are differentially coupled in epileptic and control rats. Neuronus IBRO & IRUN Neuroscience Forum. 17 – 19 April 2015. Kraków, Poland. Smyk MK, van Luijtelaar G, Drinkenburg WH. 2013. Old rhythm new methodology: differential organization of sleep-wake states in the light and the dark phase. 11th International Congress of the Polish Neuroscience Society. 15 – 17 September 2013. Poznań, Poland. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2013. Epileptic clock: circadian control of absence seizures. Understanding the neural basis of diurnality. 26 – 28 November 2013. Strasbourg, France. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2011. Do not mess with your biological clock: Seizure aggravation after the phase shift. 3rd World Congress of Chronobiology. 5 – 9 May 2011. Puebla de Zaragoza, Mexico. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2011. Do not mess with your biological clock: Seizure aggravation after the phase shift. 8th IBRO World Congress of Neuroscience. 14 – 18 July 2011. Florence, Italy. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2011. The jet lag syndrome in epileptic rats: seizures response to 8 hr phase-shift. 10th International Congress of the Polish Neuroscience Society. 21 – 24 September 2011. Łódź, Poland. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2010. The circadian control of absence seizures. 7th FENS Forum of European Neuroscience. 3 – 7 July 2010. Amsterdam, the Netherlands. Smyk MK, Coenen AML, Lewandowski MH, van Luijtelaar G. 2009. Seizure control in free-running rats. 9th International Congress of the Polish Neuroscience Society. 9 – 12 September 2009, Warsaw, Poland. 234 Acknowledgements Success has many fathers (we won’t talk about failure here! ;)). So true. I wouldn’t have reached so much without generous support from many great people. Let me express my gratitude to everybody who accompanied me during this long, difficult but also very exciting journey called the doctoral project. At first I would like to thank my supervisors. I had the pleasure to work with four brilliant gentlemen, two of them being my promoters. Some people may find it difficult, but personally, I liked it a lot. Dear Gilles, remember our first meeting? Because of the carnival you had to wait for me at the train station more than an hour. As it appeared later, that was not the last time you had to wait. You’re the most patient and understanding boss I’ve ever had. Thank you for the time we spent on designing experiments, discussing results and papers. You taught me that there’s no such thing as a bad result, that there’s no need to spend ages (really ages) on data analysis and that my graphs won’t be more beautiful because they are already perfect. You’re a word champion and a gold medalist in fighting with my stubbornness, which is very impressive because it’s a heavyweight category. Thank you for your faith and trust in me. It’s you who made me believe I can send a paper to Epilepsia just like that. From that moment I started shyly thinking about myself as a scientist. During last Neuronus party somebody said “I want to be Gilles”. I’m not sure whether the person meant your scientific achievements, dancing skills, or both. Doesn’t matter, I want to be Gilles too! Dear Marian, I said once that if I could choose a father, that would be you. I keep in mind all of your advice but the one I like the most is “Can you count Magda? Count on yourself!” I particularly like it because, when I think about all these years in our department, the advice is rubbish. I could always count on you! I really appreciate your 235 constant belief in me. Thank you for your support, encouragement to apply for any scholarship or scientific school possible, for zillion of recommendation letters you wrote for me and for a lesson how to cope in the administrative jungle (that’s my second favorite piece of advice). Thank you for all positive kicks and motivation. You’re a real chronobiologist, their timing was just perfect. Speaking of chronobiology, looking at absence epilepsy from this point of view was a fascinating adventure which wouldn’t be possible without your background. I still remember your lectures for the first year students. I thought then, oh my God I wish I could lecture like that one day. Dear Ton, this project wouldn’t be possible without you. Your warm feelings for Krakow and a friendship with Prof. Jan Kaiser let us meet and discuss whether and why chronobiology of absence epilepsy is worthy to investigate. Thank you for organizing things. Thank you for the critical reviews of the manuscripts and perseverance in a struggle with chaos inside. Thank you for all these questions “why” and “how”. You made me improve the way of selling ideas to make them understandable for the audience. Thank you for introducing me to the cool young psychologists with whom I shared a building in Krakow but surprisingly, I’ve never talked to before. With some of them, namely Wiola Walentowska, Mirek Wyczesany and Kuba Szewczyk I lived an exciting life of an external PhD student of the Radboud University Nijmegen. Dear Pim, I’m very grateful for the Janssen opportunity and your support during my stay in Beerse. I’m truly amazed how you cope with this crazy amount of work. I appreciate even more that you found time for our project. It was a great experience to be a part of your team. Please pass my kind regards to the members of the Department of Neuroscience, Janssen R&D I had the pleasure to work with. Dear Saskia (Menting-Hermeling), Heidi (Huysmans) and Hans (Krijnen), thank you for your help during preparation and conducting 236 of the experiments. I wouldn’t manage without your excellent skills. Big thank you and a hug ;) go to technicians-magicians: Gerard van Oijen, Pascal de Water and Dirk Nuyts making incredible things with the equipment and software, things I will never fully understand. Dear Family, you have never had a choice – you must love me form the definition ;). I’m pretty aware, however, how difficult it is. I would like to thank you here for all this love, patience and understanding. Agnieszka, Lucjan, Ola, Halina and Marian, during my doctoral studies you witnessed a wide range of extreme behaviors: from insane happiness to furious anger and hopeless depression. You may think I’ve chosen a difficult path. I have. Science is not a piece of cake (right uncle engineer?), but thanks to you I know who I am and I know I will always manage. Thank you for the freedom of choice and acceptance for all my decisions, especially those in opposition to what you believe in. Thank you for letting me making my own mistakes and learning by that. Thank you for being there for me. Always. My dear girls: Becia, MES and Pati (alphabetical order, I love you equally), as young people are saying nowadays, you’re my BFFs. Thank you for your presence, your patience, kindness, for wild parties and the ocean of alcohol we had together. We girls love to talk a lot. Thank you for all discussions, especially those in which we differed substantially. Of course, I appreciate gossiping too. It’s your super power to deal with me when I’m so annoying that even I can’t stand myself. I’m impressed by your threshold of tolerance. Having such friends as three of you is a privilege – it’s good to be me :). Dear Dirk, time flies but I still remember my first 2 – 3 years in Nijmegen. What a wonderful, intense time. Thank you for showing me some cool spots in the Netherlands and for giving me the best impression of a Dutch gentleman. You were always very supportive and you had a great faith in me. It meant a lot. 237 Pati and Dirk, thank you for standing next to me during the defence. Right people at the right place – I couldn’t think of anybody else. Dear Margo, Emanuel, Valerio, Serhiy, Rustam, Jeroen - my lovely Nijmegen team, thank you for the time we spent together, all these crazy moments I will remember for the rest of my life. You made my stay in the city truly awesome! What I appreciate the most is your open mind and a shoulder always ready for me to cry on. Margo, I’m pretty sure you’re well aware of your emotional support but do you know about your contribution to the thesis? Let me explain. Every time I started writing (it’s not my favorite thing to do) I heard our conversation: - I wrote an introduction! - Congrats Margo! - No, no… The word “introduction” This made the whole process a little bit more tolerable :). Dear Gilles’ Angels: Annika, Lili, Mehrnoush and Valerio. It was great to be a part of the team. Thank you for sharing an office, ideas, technical know-how, tons of chocolate, cookies and sour candies. I miss the moments we laugh so hard that the only thing we could do afterwards was going home – any productive work was impossible. Dear Biological Psychologists from Nijmegen, I wish I could teleport myself to your coffee table every day. Thank you for a warm welcome every time I came to Donders. I appreciate friendly atmosphere and invaluable help regarding work-related and non-workrelated matters. Dear Neurophysiologists and Chronobiologist from Krakow. It was, and still, it is an honour to share a working space with you. Our lab is a special place, as other jealous departments claim. It’s because of you creating this great atmosphere with a perfect balance between 238 science and social activities. With you success tastes better, failures hurt less. Dear Huber and Lagrange Family, thank you for making me feel at home during my stay in Nijmegen and Vosselaar. Being abroad alone was not that difficult knowing that you’re around. Thank you for the understanding for a Polish girl who, apart from a hard work of course, was partying a lot, sleeping till midday, drinking tea instead of water and was having problems with using a garage door to put her bike inside ;). Last but not least, Mr. Moś and Zdzisława, thank you guys for showing me that life is not only about doing experiments and writing papers. Ordinary things such as a full fridge, soft bed and willing stroking hand matter as well. Thanks for showing me in practice how the food-entrainable oscillator works and how the social cues set the biological clock. That’s the real life science! Podziękowania Sukces ma wielu ojców (nie będziemy wspominać tutaj o porażce! ;)). Jakie to prawdziwe. Nie osiągnęłabym tyle bez hojnego wsparcia wielu wspaniałych osób. Chciałabym wyrazić swoją wdzięczność wszystkim, którzy towarzyszyli mi w tej długiej, trudnej, ale i niezwykle fascynującej drodze zwanej studiami doktoranckimi. Na początku chciałabym podziękować moim przełożonym. Miałam przyjemność pracować z czterema genialnymi dżentelmenami, z których dwaj to moi promotorzy. Niektórzy sądzą, iż posiadanie tylu szefów jest męczące, aczkolwiek, osobiście bardzo to lubiłam. Drogi Gilles’u pamiętasz nasze pierwsze spotkanie? Z powodu karnawału musiałeś czekać na mnie na dworcu ponad godzinę. Jak się później okazało, nie był to ostatni raz, kiedy musiałeś na mnie czekać. Jesteś najbardziej cierpliwym i wyrozumiałym 239 szefem, jakiego kiedykolwiek miałam. Dziękuję Ci za czas poświęcony na planowanie eksperymentów, omawianie wyników i manuskryptów. Nauczyłeś mnie, że nie ma czegoś takiego jak zły wynik, że nie ma potrzeby spędzania wieków (naprawdę wieków) nad analizą danych i że moje wykresy nie będą jeszcze piękniejsze, bo już są perfekcyjne. Jesteś mistrzem świata i złotym medalistą w walce z moim uporem, co imponuje tym bardziej, iż jest to waga ciężka. Dziękuję za wiarę we mnie i za zaufanie. To Ty sprawiłeś, że uwierzyłam, że mogę wysłać papier do Epilepsji – tak po prostu. Od tego momentu zaczęłam nieśmiało postrzegać siebie, jako naukowca. Podczas ostatniej imprezy Neuronusowej ktoś powiedział „chcę być Gilles’em”. Nie jestem pewna czy osoba ta miała na myśli Twoje osiągnięcia naukowe, umiejętności taneczne czy obie te rzeczy naraz. To bez znaczenia, ja też chcę być Gilles’em! Drogi Marianie, powiedziałam kiedyś, że gdybym mogła wybrać sobie tatę to byłby to Pan. Przechowuję w pamięci wszystkie dobre rady, które od Pana dostałam, ale moją ulubioną jest ta: „Umiesz liczyć Magda? Licz na siebie!”. Lubię ją szczególnie, ponieważ gdy spoglądam na te wszystkie lata spędzone w naszym zakładzie, ta rada to wierutna bzdura. Zawsze mogłam liczyć na Pana! Naprawdę doceniam Pana niezmienną wiarę we mnie. Dziękuję za wsparcie, za zachętę do aplikowania o wszystkie możliwe stypendia i szkoły naukowe, za zyliony listów rekomendacyjnych, które Pan dla mnie napisał i za lekcję jak przetrwać w biurokratycznej dżungli (to moja druga ulubiona rada). Dziękuje za pozytywne kopy i motywację. Prawdziwy z Pana chronobiolog, zachęta zawsze przychodziła w odpowiednim czasie. Mówiąc o chronobiologii, badanie napadów nieświadomości pod tym kątem nie byłoby możliwe bez Pana naukowych korzeni. Do dzisiaj pamiętam Pana wykład dla studentów pierwszego roku. Pomyślałam sobie wtedy: Boże, chciałabym tak kiedyś wykładać. 240 Drogi Tonie, ta praca nie doszłaby do skutku bez Twojego wkładu. Twoja sympatia do Krakowa i przyjaźń z profesorem Janem Kaiserem pozwoliły nam się spotkać i przedyskutować czy i dlaczego warto badać napady nieświadomości pod kątem chronobiologicznym. Dziękuję Ci za zorganizowanie wszystkiego. Dziękuję za krytyczne recenzje naszych manuskryptów i niezłomną walkę z chaosem w nich zawartym. Dziękuję za te wszystkie pytania „dlaczego” i „jak”. Dzięki Tobie nauczyłam się lepiej „sprzedać” swoje pomysły i wyniki słuchaczom i czytelnikom. Dziękuję za przedstawienie mnie fajowym młodym psychologom, z którymi dzieliłam budynek w Krakowie, a z którymi, nie wiedzieć czemu, nie zamieniłam wcześniej ani słowa. Z niektórymi z nich, a mianowicie z Wiolą Walentowską, Mirkiem Wyczesanym i Kubą Szewczykiem dzieliłam potem żywot studenta Uniwersytetu Radboud w Nijmegen. Drogi Pim’ie jestem bardzo wdzięczna za szansę pobytu w Janssen’ie oraz Twoje wsparcie. Jestem pełna podziwu dla sposobu, w jakim radzisz sobie z tym szalonym nawałem obowiązków. Jeszcze bardziej doceniam więc, że znalazłeś czas na nasz projekt. Bycie częścią Twojego zespołu to wspaniałe doświadczenie. Proszę, przekaż moje ukłony członkom Department of Neuroscience, Janssen R&D, z którymi miałam przyjemność pracować. Droga Saskia (Menting-Hermeling), Heidi (Huysmans) oraz Hans’ie (Krijnen) dziękuję za pomoc w przygotowaniu i przeprowadzeniu doświadczeń. Nie dałabym sobie rady bez waszych niesamowitych umiejętności. Duże „dziękuję” oraz uściski wędrują do moich techników-magików: Gerard’a van Oijen, Pascal’a de Water i Dirk’a Nuyts, którzy wyczyniali ze sprzętem i oprogramowaniem takie cuda, jakich nigdy do końca nie ogarnę. Droga Rodzino, nigdy nie miałaś wyboru – musisz mnie kochać z definicji ;). Jestem jednakże świadoma, jak trudne to zadanie. Z tego miejsca chciałabym podziękować Wam za tą całą miłość, cierpliwość i 241 zrozumienie, którymi mnie obdarzyliście. Aguniu, Lucjanie, Olusiu, ciociu Halinko i wujku Marianie, podczas moich studiów doktoranckich dane Wam zachowań: było od zaobserwować szalonego szeroką szczęścia przez gamę ekstremalnych wściekły gniew do beznadziejnej depresji. Możecie pomyśleć, że wybrałam trudną drogę. Wybrałam. Nauka to nie bułka z masłem (prawda Wujku Inżynierze?), ale dzięki Wam wiem, kim jestem i wiem, że zawsze dam sobie radę. Dziękuję za wolność wyboru i akceptację dla wszystkich moich wyborów, szczególnie dla tych, z którymi Wam nie po drodze. Dziękuję za prawo do popełniania swoich własnych błędów i za naukę z nich płynącą. Dziękuję, że jesteście przy mnie. Zawsze. Moje drogie dziewczęta: Beciu, MES i Pati (kolejność alfabetyczna – kocham Was jednakowo), jak to mawia dzisiejsza młodzież, jesteście moimi BFF. Dziękuję za Waszą obecność, cierpliwość i dobroć, za dzikie melanże i ocean alkoholu wypity razem. My dziewczyny uwielbiamy dużo mówić. Dziękuję za wszystkie rozmowy, a szczególnie te, w których znacznie się różniłyśmy. Oczywiście, doceniam i ploteczki. Posiadacie niesamowitą super moc radzenia sobie ze mną w momentach, w których nawet ja sama nie mogę siebie znieść. Posiadanie takich przyjaciółek jak Wy to zaszczyt – dobrze jest być mną :). Drogi Dirk’u, czas mija nieubłaganie, ale nadal pamiętam moje 2 - 3 pierwsze lata w Nijmegen. Cóż za wspaniały, intensywny czas. Dziękuję za pokazanie mi kilku fajowych miejsc w Holandii oraz za najlepsze wrażenie odnośnie holenderskich dżentelmenów. Zawsze mnie wspierałeś i zawsze we mnie wierzyłeś. To wiele dla mnie znaczyło. Pati i Dirk’u dziękuję za towarzyszenie mi podczas obrony. Właściwi ludzie na właściwym miejscu – nie mogłam pomyśleć o nikim innym. 242 Droga Margo, Emanuel’u Valerio, Serhiy, Rustam’ie i Jeroen’ie – moja kochana drużyno z Nijmegen, dziękuję za wspólny czas i te wszystkie szalone momenty, które pozostaną ze mną do końca życia. Uczyniliście mój pobyt w mieście naprawdę odlotowym. Szczególnie doceniam Wasze otwarte umysły i ramiona zawsze gotowe, aby się na nich wypłakać. Margo, zapewne zdajesz sobie sprawę ze swojego wkładu emocjonalnego, ale czy wiesz jak przyczyniłaś się do powstania tej pracy? Za każdym razem, kiedy rozpoczynałam pisanie (co, jak wiadomo, nie jest moją ulubioną czynnością) słyszałam naszą rozmowę: - Napisałam wstęp! - Gratulacje Margo! - nie, nie… słowo „wstęp” Dzięki temu proces pisania stawał się odrobinę bardziej znośny. Drogie Aniołki Gilles’a: Annika, Lili, Mehrnoush i Valerio. Wspaniale było być częścią naszego teamu. Dziękuję za dzielenie pokoju, pomysłów, technicznego know-how, ton czekolady, ciastek i kwaśnych cukierków. Tęsknię za momentami, w których śmialiśmy się tak mocno, że musieliśmy iść do domów, bo jakakolwiek praca była niemożliwa. Drodzy Biopsycholodzy z Nijmegen, żałuję, że nie mogę teleportować się do Waszego stolika kawowego codziennie. Dziękuję za ciepłe powitania z okazji moich wizyt w Donders’ie. Doceniam przyjazną atmosferę i nieocenioną pomoc w sprawach zawodowych jak również tych poza zawodowych. Drodzy Neurofizjolodzy i Chronobiolodzy z Krakowa, zaszczytem było, i jest nadal, dzielenie z Wami miejsca pracy. Jak potwierdzają inne zazdrosne zakłady, nasz lab to szczególne miejsce. Szczególne, ponieważ to właśnie Wy tworzycie niepowtarzalny klimat z idealną równowagą pomiędzy nauką a życiem towarzyskim. Z Wami sukces smakuje lepiej, porażki bolą mniej. 243 Droga rodzino Huber i Lagrange, dzięki Wam czułam się w Nijmegen i Vosselaar jak w domu. Samotny pobyt za granicą nie był taki straszny wiedząc, że jesteście w pobliżu. Dziękuję za wyrozumiałość dla polskiej dziewczyny, która poza ciężką pracą oczywiście, dużo imprezowała, spała do południa, piła herbatę zamiast wody do obiadu i miała problemy z otwieraniem drzwi garażowych, aby zaparkować rower w środku ;). Ostatni, ale nie mniej ważni, Panie Mosiu i Zdzisławo, dziękuję Wam za pokazanie mi, iż życie nie polega jedynie na przeprowadzaniu doświadczeń i pisaniu artykułów. Zwykłe, codzienne rzeczy takie jak pełna lodówka, wygodne łóżko i ręka chętna do głaskania są równie ważne. Dzięki za pokazanie mi w praktyce jak działa oscylator nastawiany pokarmem i jak bodźce socjalne nastawiają zegar biologiczny. To są prawdziwe nauki przyrodnicze! 244 Donders Graduate School for Cognitive Neuroscience Series 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. Van Aalderen-Smeets, S.I. (2007). Neural dynamics of visual selection. Maastricht University, Maastricht, the Netherlands. Schoffelen, J.M. (2007). Neuronal communication through coherence in the human motor system. Radboud University Nijmegen, Nijmegen, the Netherlands. De Lange, F.P. (2008). Neural mechanisms of motor imagery. Radboud University Nijmegen, Nijmegen, the Netherlands. Grol, M.J. (2008). Parieto-frontal circuitry in visuomotor control. Utrecht University, Utrecht, the Netherlands. Bauer, M. (2008). Functional roles of rhythmic neuronal activity in the human visual and somatosensory system. Radboud University Nijmegen, Nijmegen, the Netherlands. Mazaheri, A. (2008). The influence of ongoing oscillatory brain activity on evoked responses and behaviour. Radboud University Nijmegen, Nijmegen, the Netherlands. Hooijmans, C.R. (2008). Impact of nutritional lipids and vascular factors in Alzheimer’s disease. Radboud University Nijmegen, Nijmegen, the Netherlands. Gaszner, B. (2008). Plastic responses to stress by the rodent urocortinergic EdingerWestphal nucleus. Radboud University Nijmegen, Nijmegen, the Netherlands. Willems, R.M. (2009). Neural reflections of meaning in gesture, language and action. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Pelt, S. (2009). Dynamic neural representations of human visuomotor space. Radboud University Nijmegen, Nijmegen, the Netherlands. Lommertzen, J. (2009). Visuomotor coupling at different levels of complexity. Radboud University Nijmegen, Nijmegen, the Netherlands. Poljac, E. (2009). Dynamics of cognitive control in task switching: Looking beyond the switch cost. Radboud University Nijmegen, Nijmegen, the Netherlands. Poser, B.A. (2009). Techniques for BOLD and blood volume weighted fMRI. Radboud University Nijmegen, Nijmegen, the Netherlands. Baggio, G. (2009). Semantics and the electrophysiology of meaning. Tense, aspect, event structure. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Wingen, G.A. (2009). Biological determinants of amygdala functioning. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Bakker, M. (2009). Supraspinal control of walking: Lessons from motor imagery. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Aarts, E. (2009). Resisting temptation: The role of the anterior cingulate cortex in adjusting cognitive control. Radboud University Nijmegen, Nijmegen, the Netherlands. 245 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. Prinz, S. (2009). Waterbath stunning of chickens – Effects of electrical parameters on the electroencephalogram and physical reflexes of broilers. Radboud University Nijmegen, Nijmegen, the Netherlands. Knippenberg, J.M.J. (2009). The N150 of the Auditory Evoked Potential from the rat amygdala: In search for its functional significance. Radboud University Nijmegen, Nijmegen, the Netherlands. Dumont, G.J.H. (2009). Cognitive and physiological effects of 3,4methylenedioxymethamphetamine (MDMA or ’ecstasy’) in combination with alcohol or cannabis in humans. Radboud University Nijmegen, Nijmegen, the Netherlands. Pijnacker, J. (2010). Defeasible inference in autism: A behavioral and electrophysiogical approach. Radboud University Nijmegen, Nijmegen, the Netherlands. De Vrijer, M. (2010). Multisensory integration in spatial orientation. Radboud University Nijmegen, Nijmegen, the Netherlands. Vergeer, M. (2010). Perceptual visibility and appearance: Effects of color and form. Radboud University Nijmegen, Nijmegen, the Netherlands. Levy, J. (2010). In cerebro unveiling unconscious mechanisms during reading. Radboud University Nijmegen, Nijmegen, the Netherlands. Treder, M. S. (2010). Symmetry in (inter)action. Radboud University Nijmegen, Nijmegen, the Netherlands. Horlings C.G.C. (2010). A weak balance: Balance and falls in patients with neuromuscular disorders. Radboud University Nijmegen, Nijmegen, the Netherlands. Snaphaan, L.J.A.E. (2010). Epidemiology of post-stroke behavioural consequences. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Dado – Van Beek, H.E.A. (2010). The regulation of cerebral perfusion in patients with Alzheimer’s disease. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Derks, N.M. (2010). The role of the non-preganglionic Edinger-Westphal nucleus in sex-dependent stress adaptation in rodents. Radboud University Nijmegen, Nijmegen, the Netherlands. Wyczesany, M. (2010). Covariation of mood and brain activity. Integration of subjective self-report data with quantitative EEG measures. Radboud University Nijmegen, Nijmegen, the Netherlands. Beurze S.M. (2010). Cortical mechanisms for reach planning. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Dijk, J.P. (2010). On the Number of Motor Units. Radboud University Nijmegen, Nijmegen, the Netherlands. Lapatki, B.G. (2010). The Facial Musculature - Characterization at a Motor Unit Level. Radboud University Nijmegen, Nijmegen, the Netherlands. Kok, P. (2010). Word order and verb inflection in agrammatic sentence production. Radboud University Nijmegen, Nijmegen, the Netherlands. 246 35. 36. 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. 51. 52. van Elk, M. (2010). Action semantics: Functional and neural dynamics. Radboud University Nijmegen, Nijmegen, the Netherlands. Majdandzic, J. (2010). Cerebral mechanisms of processing action goals in self and others. Radboud University Nijmegen, Nijmegen, the Netherlands. Snijders, T.M. (2010). More than words - Neural and genetic dynamics of syntactic unification. Radboud University Nijmegen, Nijmegen, the Netherlands. Grootens, K.P. (2010). Cognitive dysfunction and effects of antipsychotics in schizophrenia and borderline personality disorder. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Nieuwenhuis, I.L.C. (2010). Memory consolidation: A process of integration – Converging evidence from MEG, fMRI and behavior. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Menenti, L.M.E. (2010). The right language: Differential hemispheric contributions to language production and comprehension in context. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Dijk, H.P. (2010). The state of the brain, how alpha oscillations shape behaviour and event related responses. Radboud University Nijmegen, Nijmegen, the Netherlands. Meulenbroek, O.V. (2010). Neural correlates of episodic memory in healthy aging and Alzheimer’s disease. Radboud University Nijmegen, Nijmegen, the Netherlands. Oude Nijhuis, L.B. (2010). Modulation of human balance reactions. Radboud University Nijmegen, Nijmegen, the Netherlands. Qin, S. (2010). Adaptive memory: Imaging medial temporal and prefrontal memory systems. Radboud University Nijmegen, Nijmegen, the Netherlands. Timmer, N.M. (2011). The interaction of heparan sulfate proteoglycans with the amyloid protein. Radboud University Nijmegen, Nijmegen, the Netherlands. Crajé, C. (2011). (A)typical motor planning and motor imagery. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Grootel, T.J. (2011). On the role of eye and head position in spatial localisation behaviour. Radboud University Nijmegen, Nijmegen, the Netherlands. Lamers, M.J.M. (2011). Levels of selective attention in action planning. Radboud University Nijmegen, Nijmegen, the Netherlands. Van der Werf, J. (2011). Cortical oscillatory activity in human visuomotor integration. Radboud University Nijmegen, Nijmegen, the Netherlands. Scheeringa, R. (2011). On the relation between oscillatory EEG activity and the BOLD signal. Radboud University Nijmegen, Nijmegen, the Netherlands. Bögels, S. (2011). The role of prosody in language comprehension: When prosodic breaks and pitch accents come into play. Radboud University Nijmegen, Nijmegen, the Netherlands. Ossewaarde, L. (2011). The mood cycle: Hormonal influences on the female brain. Radboud University Nijmegen, Nijmegen, the Netherlands. 247 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. Kuribara, M. (2011). Environment-induced activation and growth of pituitary melanotrope cells of Xenopus laevis. Radboud University Nijmegen, Nijmegen, the Netherlands. Helmich, R.C.G. (2011). Cerebral reorganization in Parkinson’s disease. Radboud University Nijmegen, Nijmegen, the Netherlands. Boelen, D. (2011). Order out of chaos? Assessment and treatment of executive disorders in brain-injured patients. Radboud University Nijmegen, Nijmegen, the Netherlands. Koopmans, P.J. (2011). fMRI of cortical layers. Radboud University Nijmegen, Nijmegen, the Netherlands. van der Linden, M.H. (2011). Experience-based cortical plasticity in object category representation. Radboud University Nijmegen, Nijmegen, the Netherlands. Kleine, B.U. (2011). Motor unit discharges - Physiological and diagnostic studies in ALS. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Paulus, M. (2011). Development of action perception: Neurocognitive mechanisms underlying children’s processing of others’ actions. Radboud University Nijmegen, Nijmegen, the Netherlands. Tieleman, A.A. (2011). Myotonic dystrophy type 2. A newly diagnosed disease in the Netherlands. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Van Leeuwen, T.M. (2011). ‘How one can see what is not there’: Neural mechanisms of grapheme-colour synaesthesia. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Tilborg, I.A.D.A. (2011). Procedural learning in cognitively impaired patients and its application in clinical practice. Radboud University Nijmegen, Nijmegen, the Netherlands. Bruinsma, I.B. (2011). Amyloidogenic proteins in Alzheimer’s disease and Parkinson’s disease: Interaction with chaperones and inflammation. Radboud University Nijmegen, Nijmegen, the Netherlands. Voermans, N. (2011). Neuromuscular features of Ehlers-Danlos syndrome and Marfan syndrome; expanding the phenotype of inherited connective tissue disorders and investigating the role of the extracellular matrix in muscle. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Reelick, M. (2011). One step at a time. Disentangling the complexity of preventing falls in frail older persons. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Buur, P.F. (2011). Imaging in motion. Applications of multi-echo fMRI. Radboud University Nijmegen, Nijmegen, the Netherlands. Schaefer, R.S. (2011). Measuring the mind’s ear: EEG of music imagery. Radboud University Nijmegen, Nijmegen, the Netherlands. 248 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. Xu, L. (2011). The non-preganglionic Edinger-Westphal nucleus: An integration center for energy balance and stress adaptation. Radboud University Nijmegen, Nijmegen, the Netherlands. Schellekens, A.F.A. (2011). Gene-environment interaction and intermediate phenotypes in alcohol dependence. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Marle, H.J.F. (2011). The amygdala on alert: A neuroimaging investigation into amygdala function during acute stress and its aftermath. Radboud University Nijmegen, Nijmegen, the Netherlands. De Laat, K.F. (2011). Motor performance in individuals with cerebral small vessel disease: An MRI study. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Mädebach, A. (2011). Lexical access in speaking: Studies on lexical selection and cascading activation. Radboud University Nijmegen, Nijmegen, the Netherlands. Poelmans, G.J.V. (2011). Genes and protein networks for neurodevelopmental disorders. Radboud University Nijmegen, Nijmegen, the Netherlands. Van Norden, A.G.W. (2011). Cognitive function in elderly individuals with cerebral small vessel disease. An MRI study. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Jansen, E.J.R. (2011). New insights into V-ATPase functioning: the role of its accessory subunit Ac45 and a novel brain-specific Ac45 paralog. Radboud University Nijmegen, Nijmegen, the Netherlands. Haaxma, C.A. (2011). New perspectives on preclinical and early stage Parkinson's disease. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Haegens, S. (2012). On the functional role of oscillatory neuronal activity in the somatosensory system. Radboud University Nijmegen, Nijmegen, the Netherlands. van Barneveld, D.C.P.B.M. (2012). Integration of exteroceptive and interoceptive cues in spatial localization. Radboud University Nijmegen, Nijmegen, the Netherlands. Spies, P.E. (2012). The reflection of Alzheimer disease in CSF. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Helle, M. (2012). Artery-specific perfusion measurements in the cerebral vasculature by magnetic resonance imaging. Radboud University Nijmegen, Nijmegen, the Netherlands. Egetemeir, J. (2012). Neural correlates of real-life joint action. Radboud University Nijmegen, Nijmegen, the Netherlands. Janssen, L. (2012). Planning and execution of (bi)manual grasping. Radboud University Nijmegen, Nijmegen, the Netherlands. 249 83. Vermeer, S. (2012). Clinical and genetic characterisation of autosomal recessive cerebellar ataxias. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. 84. Vrins, S. (2012). Shaping object boundaries: Contextual effects in infants and adults. Radboud University Nijmegen, Nijmegen, the Netherlands. Weber, K.M. (2012). The language learning brain: Evidence from second language and bilingual studies of syntactic processing. Radboud University Nijmegen, Nijmegen, the Netherlands. Verhagen, L. (2012). How to grasp a ripe tomato. Utrecht University, Utrecht, the Netherlands. Nonkes, L.J.P. (2012). Serotonin transporter gene variance causes individual differences in rat behaviour: For better and for worse. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. 85. 86. 87. 88. 89. 90. 91. 92. 93. 94. 95. 96. 97. 98. Joosten-Weyn Banningh, L.W.A. (2012). Learning to live with Mild Cognitive Impairment: development and evaluation of a psychological intervention for patients with Mild Cognitive Impairment and their significant others. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Xiang, HD. (2012). The language networks of the brain. Radboud University Nijmegen, Nijmegen, the Netherlands. Snijders, A.H. (2012). Tackling freezing of gait in Parkinson's disease. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Rouwette, T.P.H. (2012). Neuropathic pain and the brain - Differential involvement of corticotropin-releasing factor and urocortin 1 in acute and chronic pain processing. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Van de Meerendonk, N. (2012). States of indecision in the brain: Electrophysiological and hemodynamic reflections of monitoring in visual language perception. Radboud University Nijmegen, Nijmegen, the Netherlands. Sterrenburg, A. (2012). The stress response of forebrain and midbrain regions: Neuropeptides, sex-specificity and epigenetics. Radboud University Nijmegen, Nijmegen, The Netherlands. Uithol, S. (2012). Representing action and intention. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Dam, W.O. (2012). On the specificity and flexibility of embodied lexicalsemantic representations. Radboud University Nijmegen, Nijmegen, The Netherlands. Slats, D. (2012). CSF biomarkers of Alzheimer’s disease: Serial sampling analysis and the study of circadian rhythmicity. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Van Nuenen, B.F.L. (2012). Cerebral reorganization in premotor parkinsonism. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. van Schouwenburg, M.R. (2012). Fronto-striatal mechanisms of attentional control. Radboud University Nijmegen, Nijmegen, The Netherlands. 250 99. 100. 101. 102. 103. 104. 105. 106. 107. 108. 109. 110. 111. 112. 113. Azar, M.G. (2012). On the theory of reinforcement learning: Methods, convergence analysis and sample complexity. Radboud University Nijmegen, Nijmegen, The Netherlands. Meeuwissen, E.B. (2012). Cortical oscillatory activity during memory formation. Radboud University Nijmegen, Nijmegen, The Netherlands. Arnold, J.F. (2012). When mood meets memory: Neural and behavioral perspectives on emotional memory in health and depression. Radboud University Nijmegen, Nijmegen, The Netherlands. Gons, R.A.R. (2012). Vascular risk factors in cerebral small vessel disease: A diffusion tensor imaging study. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Wingbermühle, E. (2012). Cognition and emotion in adults with Noonan syndrome: A neuropsychological perspective. Radboud University Nijmegen, Nijmegen, The Netherlands. Walentowska, W. (2012). Facing emotional faces. The nature of automaticity of facial emotion processing studied with ERPs. Radboud University Nijmegen, Nijmegen, The Netherlands. Hoogman, M. (2012). Imaging the effects of ADHD risk genes. Radboud University Nijmegen, Nijmegen, The Netherlands. Tramper, J. J. (2012). Feedforward and feedback mechanisms in sensory motor control. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Eijndhoven, P. (2012). State and trait characteristics of early course major depressive disorder. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Visser, E. (2012). Leaves and forests: Low level sound processing and methods for the large-scale analysis of white matter structure in autism. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Tooren-Hoogenboom, N. (2012). Neuronal communication in the synchronized brain. Investigating the functional role of visually-induced gamma band activity: Lessons from MEG. Radboud University Nijmegen, Nijmegen, The Netherlands. Henckens, M.J.A.G. (2012). Imaging the stressed brain. Elucidating the time- and region-specific effects of stress hormones on brain function: A translational approach. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Kesteren, M.T.R. (2012). Schemas in the brain: Influences of prior knowledge on learning, memory, and education. Radboud University Nijmegen, Nijmegen, The Netherlands. Brenders, P. (2012). Cross-language interactions in beginning second language learners. Radboud University Nijmegen, Nijmegen, The Netherlands. Ter Horst, A.C. (2012). Modulating motor imagery. Contextual, spatial and kinaesthetic influences. Radboud University Nijmegen, Nijmegen, The Netherlands. 251 114. Tesink, C.M.J.Y. (2013). Neurobiological insights into language comprehension in 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. 129. autism: Context matters. Radboud University Nijmegen, Nijmegen, The Netherlands. Böckler, A. (2013). Looking at the world together. How others’ attentional relations to jointly attended scenes shape cognitive processing. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Dongen, E.V. (2013). Sleeping to Remember. On the neural and behavioral mechanisms of sleep-dependent memory consolidation. Radboud University Nijmegen, Nijmegen, The Netherlands. Volman, I. (2013). The neural and endocrine regulation of emotional actions. Radboud University Nijmegen, Nijmegen, The Netherlands. Buchholz, V. (2013). Oscillatory activity in tactile remapping. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Deurzen, P.A.M. (2013). Information processing and depressive symptoms in healthy adolescents. Radboud University Nijmegen, Nijmegen, The Netherlands. Whitmarsh, S. (2013). Nonreactivity and metacognition in mindfulness. Radboud University Nijmegen, Nijmegen, The Netherlands. Vesper, C. (2013). Acting together: Mechanisms of intentional coordination. Radboud University Nijmegen, Nijmegen, The Netherlands. Lagro, J. (2013). Cardiovascular and cerebrovascular physiological measurements in clinical practice and prognostics in geriatric patients. Radboud University Nijmegen Medical Centre, Nijmegen, the Netherlands. Eskenazi, T.T. (2013). You, us & them: From motor simulation to ascribed shared intentionality in social perception. Radboud University Nijmegen, Nijmegen, The Netherlands. Ondobaka, S. (2013). On the conceptual and perceptual processing of own and others’ behavior. Radboud University Nijmegen, Nijmegen, The Netherlands. Overvelde, J.A.A.M. (2013). Which practice makes perfect? Experimental studies on the acquisition of movement sequences to identify the best learning condition in good and poor writers. Radboud University Nijmegen, Nijmegen, The Netherlands. Kalisvaart, J.P. (2013). Visual ambiguity in perception and action. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Kroes, M. (2013). Altering memories for emotional experiences. Radboud University Nijmegen, Nijmegen, The Netherlands. Duijnhouwer, J. (2013). Studies on the rotation problem in self-motion perception. Radboud University Nijmegen, Nijmegen, The Netherlands. Nijhuis, E.H.J (2013). Macroscopic networks in the human brain: Mapping connectivity in healthy and damaged brains. University of Twente, Enschede, The Netherlands 252 130. Braakman, M. H. (2013). Posttraumatic stress disorder with secondary psychotic 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 143. 144. features. A diagnostic validity study among refugees in the Netherlands. Radboud University Nijmegen, Nijmegen, The Netherlands. Zedlitz, A.M.E.E. (2013). Brittle brain power. Post-stroke fatigue, explorations into assessment and treatment. Radboud University Nijmegen, Nijmegen, The Netherlands. Schoon, Y. (2013). From a gait and falls clinic visit towards self-management of falls in frail elderly. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Jansen, D. (2013). The role of nutrition in Alzheimer's disease - A study in transgenic mouse models for Alzheimer's disease and vascular disorders. Radboud University Nijmegen, Nijmegen, The Netherlands. Kos, M. (2013). On the waves of language - Electrophysiological reflections on semantic and syntactic processing. Radboud University Nijmegen, Nijmegen, The Netherlands. Severens, M. (2013). Towards clinical BCI applications: Assistive technology and gait rehabilitation. Radboud University Nijmegen, Nijmegen, Sint Maartenskliniek, Nijmegen, The Netherlands. Bergmann, H. (2014). Two is not always better than one: On the functional and neural (in)dependence of working memory and long-term memory. Radboud University Nijmegen, Nijmegen, The Netherlands. Wronka, E. (2013). Searching for the biological basis of human mental abilitites. The relationship between attention and intelligence studied with P3. Radboud University Nijmegen, Nijmegen, The Netherlands. Lüttjohann, A.K. (2013). The role of the cortico-thalamo-cortical system in absence epilepsy. Radboud University Nijmegen, Nijmegen, The Netherlands. Brazil, I.A. (2013). Change doesn’t come easy: Dynamics of adaptive behavior in psychopathy. Radboud University Nijmegen, Nijmegen, The Netherlands. Zerbi, V. (2013). Impact of nutrition on brain structure and function. A magnetic resonance imaging approach in Alzheimer mouse models. Radboud University Nijmegen, Nijmegen, The Netherlands. Delnooz, C.C.S. (2014). Unravelling primary focal dystonia. A treatment update and new pathophysiological insights. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Bultena, S.S. (2013). Bilingual processing of cognates and language switches in sentence context. Radboud University Nijmegen, Nijmegen, The Netherlands. Janssen, G. (2014). Diagnostic assessment of psychiatric patients: A contextual perspective on executive functioning. Radboud University Nijmegen, Nijmegen, The Netherlands. Piai, V. Magalhães (2014). Choosing our words: Lexical competition and the involvement of attention in spoken word production. Radboud University Nijmegen, Nijmegen, The Netherlands. 253 145. Van Ede, F. (2014). Preparing for perception. On the attentional modulation, 146. 147. 148. 149. 150. perceptual relevance and physiology of oscillatory neural activity. Radboud University Nijmegen, Nijmegen, The Netherlands. Brandmeyer, A. (2014). Auditory perceptual learning via decoded EEG neurofeedback: a novel paradigm. Radboud University Nijmegen, Nijmegen, The Netherlands. Radke, S. (2014). Acting social: Neuroendocrine and clinical modulations of approach and decision behavior. Radboud University Nijmegen, Nijmegen, The Netherlands. Simanova, I. (2014). In search of conceptual representations in the brain: towards mind-reading. Radboud University Nijmegen, Nijmegen, The Netherlands. Kok, P. (2014). On the role of expectation in visual perception: A top-down view of early visual cortex. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Geldorp, B. (2014). The long and the short of memory: Neuropsychological studies on the interaction of working memory and long-term memory formation. Radboud University Nijmegen, Nijmegen, The Netherlands. 151. Meyer, M. (2014). The developing brain in action - Individual and joint action 152. 153. 154. 155. 156. 157. 158. 159. 160. processing. Radboud University Nijmegen, Nijmegen, The Netherlands. Wester, A. (2014). Assessment of everyday memory in patients with alcohol-related cognitive disorders using the Rivermead Behavioural Memory Test. Radboud University Nijmegen, Nijmegen, The Netherlands. Koenraadt, K. (2014). Shedding light on cortical control of movement. Radboud University Nijmegen, Nijmegen; Sint Maartenskliniek, Nijmegen, The Netherlands. Rutten-Jacobs, L.C.A. (2014). Long-term prognosis after stroke in young adults. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Herbert, M.K. (2014). Facing uncertain diagnosis: the use of CSF biomarkers for the differential diagnosis of neurodegenerative diseases. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Llera Arenas, A. (2014). Adapting brain computer interfaces for non-stationary changes. Radboud University Nijmegen, Nijmegen, The Netherlands. Smulders, K. (2014). Cognitive control of gait and balance in patients with chronic stroke and Parkinson's disease. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Boyacioglu, R. (2014). On the application of ultra-fast fMRI and high resolution multiband fMRI at high static field strengths. Radboud University Nijmegen, Nijmegen, The Netherlands. Kleinnijenhuis, M. (2014). Imaging fibres in the brain. Radboud University Nijmegen, Nijmegen, The Netherlands. Geuze, J. (2014). Brain Computer Interfaces for Communication: Moving beyond the visual speller. Radboud University Nijmegen, Nijmegen, The Netherlands. 254 161. Platonov, A. (2014). Mechanisms of binocular motion rivalry. Radboud University Nijmegen, Nijmegen, The Netherlands. 162. Van der Schaaf, M.E. (2014). Dopaminergic modulation of reward and punishment 163. 164. 165. 166. 167. 168. 169. 170. 171. 172. 173. 174. 175. 176. learning. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Aerts, M.B. (2014). Improving diagnostic accuracy in parkinsonism. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Vlek, R. (2014). From Beat to BCI: A musical paradigm for, and the ethical aspects of Brain-Computer Interfacing. Radboud University Nijmegen, Nijmegen, The Netherlands. Massoudi, R. (2014). Interaction of task-related and acoustic signals in single neurons of monkey auditory cortex. Radboud University Nijmegen, Nijmegen, The Netherlands Stolk, A. (2014). On the generation of shared symbols. Radboud University Nijmegen, Nijmegen, The Netherlands. Krause F. (2014). Numbers and magnitude in the brain: A sensorimotor grounding of numerical cognition. Radboud University Nijmegen, Nijmegen, The Netherlands. Munneke, M.A.M. (2014). Measuring and modulating the brain with non-invasive stimulation. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Von Borries, K. (2014). Carrots & Sticks - a neurobehavioral investigation of affective outcome processing in psychopathy. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Meel-van den Abeelen, A.S.S. (2014). In control. Methodological and clinical aspects of cerebral autoregulation and haemodynamics. Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands. Leoné, F.T.M. (2014). Mapping sensorimotor space: Parieto-frontal contributions to goal-directed movements. Radboud University Nijmegen, Nijmegen, The Netherlands. Van Kessel, M. (2014). Nothing left? How to keep on the right track - Spatial and non-spatial attention processes in neglect after stroke. Radboud University, Nijmegen, The Netherlands. Vulto-van Silfhout, A. T. (2014). Detailed, standardized and systematic phenotyping for the interpretation of genetic variation. Radboud University Medical Centre, Nijmegen, The Netherlands. Arnoldussen, D. (2015). Cortical topography of self-motion perception. Radboud University, Nijmegen, The Netherlands. Meyer, M.C. (2015). “Inbetween Modalitiers combined EEG – fMR”. Radboud University, Nijmegen, The Netherlands. Bralten, J. (2015). Genetic factors and the brain in ADHD. Radboud university medical center, Nijmegen, The Netherlands. 255 177. Spaak, E. (2015). On the role of alpha oscillations in structuring neural information processing. Radboud University, Nijmegen, The Netherlands. 178. Van der Doelen, R. (2015). Translational psychiatry; the twists and turns of early life 179. 180. 181. 182. 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. stress and serotonin transporter gene variation. Radboud university medical center, Nijmegen, The Netherlands. Lewis, C. (2015). The structure and function of intrinsic brain activity. Radboud University, Nijmegen, The Netherlands. Huang, Lili. (2015). The subiculum: a promising new target of deep brain stimulation in temporal lobe epilepsy. Investigation of closed-loop and open-loop high frequency stimulation of the subiculum in seizure and epilepsy models in rats. Radboud University, Nijmegen, The Netherlands. Maaijwee, N.A.M.M (2015). Long-term neuropsychological and social consequences after stroke in young adults. Radboud university medical center, Nijmegen, The Netherlands. Meijer, F.J.A. (2015). Clinical Application of Brain MRI in Parkinsonism: From Basic to Advanced Imaging. Radboud university medical center, Nijmegen, The Netherlands. Van der Meij, R. (2015). On the identification, characterization and investigation of phase dependent coupling in neuronal networks. Radboud University, Nijmegen, The Netherlands. Todorovic, A. (2015). Predictive adaptation in early auditory processing. Radboud University, Nijmegen, The Netherlands. Horschig, J.M. (2015). Flexible control and training of posterior alpha-band oscillations. Radboud University, Nijmegen, The Netherlands. Vogel, S. (2015). The runner-up: on the role of the mineralocorticoid receptor in human cognition. Radboud University Medical Center, Nijmegen, The Netherlands. Clemens, I.A.H. (2015). Multisensory integration in orientation and self-motion perception. Radboud University, Nijmegen, The Netherlands. Nonnekes, J. (2015). Balance and gait in neurodegenerative disease: what startle tells us about motor control. Radboud university medical center, Nijmegen, The Netherlands. Stapel, J.C. (2015). Action prediction and the development thereof. Radboud University, Nijmegen, The Netherlands. De Grauwe, S. (2015). The Processing of Derivations in Native and Non-Native Speakers of Dutch. Radboud University, Nijmegen, The Netherlands. Atucha Treviño, E. (2015). Emotional Modulation of Memory: Mechanisms underlying strength and accuracy of Memory. Radboud University, Nijmegen, The Netherlands Bosch, M.P.C. (2015) Needles on the Couch; acupuncture in the Treatment of Depression, Schizophrenia and Sleep Disorders. Radboud University, Nijmegen, The Netherlands. Marshall, T. (2015) On the Control and manipulation of Alpha and Gamma oscillations in visual cortex. Radboud University, Nijmegen, The Netherlands. 256 194. Rijken, N. (2015). Balance and gait in facioscapulohumeral muscular dystrophy, relations with individual muscle involvement. Radboud University Medical Center, Nijmegen, The Netherlands. 195. Blokpoel, M. (2015). Understanding understanding: A computational-level perspective. Radboud University, Nijmegen, The Netherlands. 196. Smyk, M. (2015). Chronobiology of absence epilepsy. Radboud University, Nijmegen, The Netherlands. 257
© Copyright 2026 Paperzz