Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 Contents lists available at SciVerse ScienceDirect Progress in Neuro-Psychopharmacology & Biological Psychiatry journal homepage: www.elsevier.com/locate/pnp Updating the mild encephalitis hypothesis of schizophrenia K. Bechter ⁎ Ulm University, Clinic for Psychiatry and Psychotherapy II, Ludwig-Heilmeyer-Str. 2, D-89312 Günzburg, Germany a r t i c l e i n f o Article history: Received 25 January 2012 Received in revised form 11 June 2012 Accepted 25 June 2012 Available online 3 July 2012 Keywords: Autoimmunity Gene–environment–immune interactions Low-level neuroinflammation Mild encephalitis hypothesis Psychoneuroimmunology Schizophrenia a b s t r a c t Schizophrenia seems to be a heterogeneous disorder. Emerging evidence indicates that low level neuroinflammation (LLNI) may not occur infrequently. Many infectious agents with low overall pathogenicity are risk factors for psychoses including schizophrenia and for autoimmune disorders. According to the mild encephalitis (ME) hypothesis, LLNI represents the core pathogenetic mechanism in a schizophrenia subgroup that has syndromal overlap with other psychiatric disorders. ME may be triggered by infections, autoimmunity, toxicity, or trauma. A ‘late hit’ and gene–environment interaction are required to explain major findings about schizophrenia, and both aspects would be consistent with the ME hypothesis. Schizophrenia risk genes stay rather constant within populations despite a resulting low number of progeny; this may result from advantages associated with risk genes, e.g., an improved immune response, which may act protectively within changing environments, although they are associated with the disadvantage of increased susceptibility to psychotic disorders. Specific schizophrenic symptoms may arise with instances of LLNI when certain brain functional systems are involved, in addition to being shaped by pre-existing liability factors. Prodrome phase and the transition to a diseased status may be related to LLNI processes emerging and varying over time. The variability in the course of schizophrenia resembles the varying courses of autoimmune disorders, which result from three required factors: genes, the environment, and the immune system. Preliminary criteria for subgrouping neurodevelopmental, genetic, ME, and other types of schizophrenias are provided. A rare example of ME schizophrenia may be observed in Borna disease virus infection. Neurodevelopmental schizophrenia due to early infections has been estimated by others to explain approximately 30% of cases, but the underlying pathomechanisms of transition to disease remain in question. LLNI (e.g. from reactivation related to persistent infection) may be involved and other pathomechanisms including dysfunction of the blood–brain barrier or the blood–CSF barrier, CNS-endogenous immunity and the volume transmission mode balancing wiring transmission (the latter represented mainly by synaptic transmission, which is often described as being disturbed in schizophrenia). Volume transmission is linked to CSF signaling; and together could represent a common pathogenetic link for the distributed brain dysfunction, dysconnectivity, and brain structural abnormalities observed in schizophrenia. In addition, CSF signaling may extend into peripheral tissues via the CSF outflow pathway along brain nerves and peripheral nerves, and it may explain the peripheral topology of neuronal dysfunctions found, like in olfactory dysfunction, dysautonomia, and even in peripheral tissues, i.e., the muscle lesions that were found in 50% of cases. Modulating factors in schizophrenia, such as stress, hormones, and diet, are also modulating factors in the immune response. Considering recent investigations of CSF, the ME schizophrenia subgroup may constitute approximately 40% of cases. © 2012 Elsevier Inc. All rights reserved. 1. Introduction Since the first version of the mild encephalitis (ME) hypothesis was proposed (Bechter, 2001), many new findings and further hypotheses have been published that fit in with or support the ME hypothesis. The ME hypothesis characterized a subgroup of severe psychiatric disorders, mainly on the affective and schizophrenic spectra, in which low level neuroinflammation (LLNI) causally underlies the disorder as the core pathogenetic mechanism. Other underlying ⁎ Tel.: +49 8221 96 2540/96 00; fax: +49 8221 96 2736. E-mail address: [email protected]. 0278-5846/$ – see front matter © 2012 Elsevier Inc. All rights reserved. doi:10.1016/j.pnpbp.2012.06.019 causes of schizophrenia that have been proposed include a Th1– Th2-imbalance and other immune abnormalities (Muller and Schwarz, 2010), cytokine abnormalities (Licinio and Wong, 1999, Maes, 1997), the two-hit-hypothesis (Bayer et al., 1999), the genetic inflammatory hypothesis (Hanson and Gottesman, 2005), the neuroprotective hypothesis (Lang et al., 2004), and the imbalance of kynurenic pathways related to neuroinflammation and neurodegeneration (Myint and Kim, 2003, Myint et al., 2011). The ME hypothesis assumed that LLNI prevailed and was important during critical time periods of disease and, although it overlaps with several psychiatric diagnostic categories, it was thought to be especially relevant in affective and schizophrenic disorders. The etiologies proposed to be involved in LLNI are varied, including infections, 72 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 autoimmunity, infection-triggered autoimmunity, toxicity (also including endogenous toxicity or disturbed protective capacity, e.g. caused by hunger), and trauma. LLNI pathomechanisms may be influenced by or even depend on pre-existing genetic factors and liabilities, including immune and inflammatory response-related genes, and the final clinical outcome may be influenced by independent factors at different pathogenetic levels, possibly relevant at multiple times over the course of the inflammation. A complex etiopathogenetic scenario is the rule rather than the exception, as demonstrated by recent insights into the infectious determinants involved in chronic CNS diseases (O'Connor et al., 2006). Several reviews of the accumulated findings on neuropsychoimmunology in schizophrenia have been published in the past two decades (Bahn and Schwarz, 2011, Licinio and Wong, 1999, Maes, 1997, Muller and Schwarz, 2010, Myint et al., 2011). The previous ME view that the diagnostic spectrum overlapped with other severe psychiatric disorders was supported by the results of large genetic studies (see below) and is consistent with longstanding knowledge about the lack of syndromal-specific etiologies in psychiatric disorders (Bechter, 2001, Gross and Huber, 2007). The ME hypothesis has recently been supported by results from epidemiological studies involving a number of infectious agents as risk factors (Benros et al., 2011, Brown et al., 2001, 2004, Dalman et al., 2008, Niebuhr et al., 2008, Yolken and Torrey, 2008) and from a large milestone study showing an additive effect of severe infections and autoimmune disorders over time (Benros et al., 2011). Findings from studies relying on newly introduced research methods such as proteomics (Holmes et al., 2006, Schwarz et al., 2012) and immune cell blood analysis (Drexhage et al., 2010a), as well as from CSF investigations (Bechter et al., 2010a, Maxeiner et al., 2009), support the view that LLNI may prevail in a considerable number of schizophrenic patients; specifically in CSF investigations, LLNI is involved in approximately 40% of cases. In this article, the ME concept is updated, and an ME subgroup comprising heterogeneous subtypes of schizophrenia is discussed. To understand the complexity of the questions involved, the definitions of inflammation must first be discussed. 2. Definitions of inflammation The preliminary definition of ME included the possibility that over short time periods, small (classical) inflammatory lesions (of microscopic size) within the CNS might prevail and might remain undetected in clinical cases due to the limitations of the methods available in the clinical approach (Table 1). LLNI pathomechanisms should generally involve brain cells, immune cells and solutes, as well as the respective exchanges between three important bodily compartments: the blood, the CSF and the CNS plus its extracellular fluid. The immune-inflammatory mediators and cells involved in LLNI may vary over time and may shape the psychiatric syndromes associated with LLNI. In the meantime, inflammation was generally redefined (see Table 2) to include a large number of possible factors on the cellular and humoral levels, the full ranges of which are only partly applicable in clinical situations. In vivo assessment and thus definition of both systemic inflammation and especially CNS inflammation remain generally limited, as the CNS is well-protected and difficult to access. Nevertheless, low-degree inflammation appears to be frequent in clinical settings, and there is little reason to assume that inflammation in the CNS occurs with any less frequency (and perhaps with greater frequency) than it does in other organs; immune regulation within the CNS is both more complex and more separated than it is in other organs, constituting an endogenous CNS immune response system (McGeer and McGeer, 1995). The roles of CSF cells were redefined in experimental neuroimmunology as playing prominent and highly active roles in the immune surveillance of the CNS during both health and disease (Schwartz and Shechter, 2010). Moreover, the CSF interacts with extracellular CNS fluid (ECF), and together, the CSF and the ECF provide signaling functions formulated in the volume transmission mode concept (Fuxe et al., 2010). These insights are corroborated by knowledge gained in the field of clinical neurology, which suggests that the investigation of the CSF is of outstanding importance for the diagnosis and thus for the pathogenesis of CNS inflammation. 2.1. Interaction between central and peripheral immunity Based on recent insights, the BBB was redefined as having two locally differing exchange components: solutes and cells (Bechmann et al., 2007). The BCB was anatomically defined as involving the choroid plexus and the circumventricular organ (Wolburg and Paulus, 2010). The choroid plexus secretes CSF and signaling molecules and is a site of active cell trafficking between the blood and the CSF (Engelhardt and Sorokin, 2009, Marques et al., 2009, Ransohoff, 2009). Modern CSF analyses have been validated by broad experience with neurological and especially neuroinflammatory disorders, leading to sophisticated rules (Reiber and Peter, 2001). However, such advanced versions of CSF analysis have only rarely been applied in psychiatric disorders thus far, and one should keep in mind that systemic inflammation and CNS-specific inflammation must be assessed in parallel. 2.2. Defining LLNI In experimental studies, LLNI-related situations are now often described (see, for example O'Callaghan et al., 2008), but a generally accepted definition is pending. Several major aspects of LLNI are briefly described here (see Table 3). The ME hypothesis (compared Table 1 The mild encephalitis (ME) hypothesis updated. • “ME is understood as a non-lethal, low grade cellular-infiltrative and/or humoral brain inflammation hypothetically underlying and basically explaining variable psychiatric symptoms/syndromes, especially a subgroup of schizophrenic or affective psychoses, possibly accompanied by neurological soft but not hard signs. Inflammatory cell infiltrates within the brain are assumed to be small and short-lived, inflammation later characterized by humoral ‘inflammatory upregulation’, which remains to be exactly defined. ME may only rarely be diagnosed by established clinical methods in the individual patient because of low sensitivity of methods” (Bechter, 2001, Neurol Psychiatry Brain Res 2001; 9:55–70) • Low grade neuroinflammation (LLNI) as major pathogenetic aspect, difficult to detect by available diagnostic methods, but core and sufficient to induce brain dysfunction resulting in psychiatric syndrome, as a diagnostic category termed ME. • Various psychiatric symptoms may be related to ME, the LLNI process may change over time (compare changing symptomatology with emerging classical encephalitis) • ME may become chronic for example in therapy-resistant affective and schizophrenic spectrum disorders • Etiologies may differ: agents (viruses, bacteria, protozoa), autoimmune, toxic, possibly traumatic injuries (including ‘endogenous’ injuries, see interaction between brain and systemic immunity) • Contributive factors in infectious ME: genetic factors, type/strain of agent, immune status, additional environmental factors • Independent liability factors: pre-existent factors (e.g. inborn, or neurodevelopmental, which may especially influence the type of syndromal outcome), changing environments, chance • Note: Course variability of schizophrenic spectrum disorders (prodromes, phases, ‘Schübe’, progression) and change of diagnosis in early years of illness might associate with emerging and changing LLNI process K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 73 Table 2 Definitons of inflammation. Classical inflammation (acc. to Celsus around AD): rubor, calor, dolor, tumor; functio laesa (last criterion added in medieval times) Conventional histopathology/virology: inflammatory cellular infiltrates within tissue visible under the microscope Recent definition (acc. to C. Nathan, 2002): “inflammation is a complex set of interactions among soluble factors and cells that can arise in any tissue in response to traumatic, infectious, post-ischaemic, toxic or autoimmune injury” Clinical definition: derived from above definitions with surprising weaknesses not least depending from diagnostic approaches applicable in vivo. Note: rather weak clinico-pathological correlations between brain inflammation and symptoms evoked. Low level neuroinflammation (LLNI) is often used as a term in experimental and now also in clinical publications (for example Keizman et al., 2009), but consensus criteria in both fields are lacking. The common aspect in such publications was that classical inflammation was not found but molecular or cellular abnormalities of minor degree, like may be found in beginning classical inflammation. The definition of ME (Table 1) remains preliminary. ME hypothesis has to be probed in psychiatric disorders with yet unknown etiopathogenesis, or in psychiatric disorders accompanying systemic inflammatory disorders, or in early or late stages of classical neuroinflammatory disorders. Recent studies in therapy-resistant affective and schizophrenic spectrum disorders performed with modern CSF analytic methods demonstrated subgroups presenting minor CSF abnormalities matching the LLNI concept (Bechter et al., 2010a, 2010b). Grading of diagnostic validity of ME: possible–probable–definite The grading should follow the accepted rules in the diagnosis of neuroinflammatory disorders, especially the CSF diagnostics (compared with Reiber and Peter 2001; Wildemann et al 2010) and neuroimaging (Osborn et al, 2010). to Bechter, 2001) has yet to be generally accepted. Compromises between clinical and theoretical approaches seem inevitable; we recall the difficulties and compromises involved in defining HIV encephalitis (HIVe). A diagnosis of HIVe required the detection of a minimum of three inflammatory lesions within the post-mortem brain, which matched well with the symptoms of clinical HIVe (Cherner et al., 2007). However, this consensus definition was considerably flawed: even one single inflammatory lesion detected under the microscope should theoretically be termed encephalitis. In addition, considering the technical details (the considerable distance in between the cuts through the brain), one can infer that many more lesions would be found in such a brain if it were studied more carefully. This compromise led to a definition that suffered from such extreme theoretical flaws that one might ask why such a compromise was deemed necessary. It has long been known that the brain can be surprisingly tolerant to pathology, and this is certainly the case in HIVe: the high cut-off required by the HIVe definition was apparently related to the low clinical symptom load observed in cases with a low number of classical inflammatory lesions; yet, this is not to say that there was an absence of inflammation, if a more basic definition of inflammation is considered. Such an interpretation is strengthened by details provided by HIVe imaging: the early detection of HIV infections associated with neuropathophysiology can be achieved with improved magnetic resonance imaging methods (i.e., with diffusion weighted imaging, but not yet with spectroscopy): unspecific imaging findings indicated that neurotoxicity is mediated by HIV infection of the tissues, and was paralleling findings in histology (Bucy et al., 2011). This type of neurotoxicity-associated tissue response apparently matches with what has been termed here as LLNI. The assumption that LLNI is currently under-diagnosed is further supported in light of changing views about other types of inflammation: one type has recently been defined as parainflammation, but many more remain unclassified as of yet (Medzhitov, 2008). 2.3. Molecular neuropathology from infections The story does not end here: infections have been shown to induce cellular changes and dysfunction even without evidence of inflammation. This phenomenon is described as the molecular anatomy of viral disease (Oldstone, 1987): viruses are able to cause disease in the absence of any morphologic evidence of cell injury (Oldstone, 1990). It is unclear whether such infection-induced molecular changes or cellular dysfunctions should be included or excluded from a definition of LLNI. Nevertheless, such molecular mechanisms should be taken into account and could be especially relevant in psychoses. Thus, we now have two undefined borders to LLNI: one of classical encephalitis and one of non-inflammatory molecular pathology after infection. If molecular pathology were to be subsumed under LLNI, would molecular LLNI (molLLNI) then be an acceptable term for it? 2.4. Proposing ME schizophrenia (meS) as a preliminary diagnostic category From this dilemma, it appears that the diagnosis of LLNI in clinical situations first requires precise observation of the basic rules established in neurology for the diagnosis of neuroinflammation (for such see Reiber and Peter, 2001, Wildemann et al., 2010). One should, however, be aware of the low sensitivity of methods for diagnosing LLNI, which need to be improved (compared with Bechter et al., 2010a). Notwithstanding these limitations in the diagnostic approach, the evidence that LLNI may prevail and be relevant in psychiatric disorders and the putative meS subgroup is considerable (Bechter et al., 2010a). Therefore, here a preliminary categorization of the meS subgroup is proposed and differentiated from other schizophrenia subgroups (see Table 4, and for explanations, see the forthcoming chapters): one might classify the various types of meS as follows: one might diagnose primary meS, in which LLNI is related Table 3 Brain barriers and CNS inflammation. The immune privilege of the brain is maintained by the brain barriers and, in contrast to previous views, by extensive immune control mechanisms within the CNS and the CSF spaces. Blood–brain barrier (BBB) and blood–CSF barrier (BCB) represent anatomically and functionally different barriers (Engelhardt and Sorokin, 2009). The BBB was dichotomized in a barrier for solutes versus cells (Bechmann et al, 2007). The anatomically defined BCB embraces the choroid plexus and circumventricular organs (Wolburg and Paulus, 2010). The term BCB dysfunction (Reiber and Peter, 2001) represents an interpretation of CSF protein abnormalities which may result from interactions mainly at the meningeal–CSF barrier and in addition at the anatomically defined BCB. The barriers between meninges and CSF spaces appears important generally but incompletely investigated and the terminology (e.g. arachnoid barrier) appears to be inconsistently used (review in Bechter et al, in prep.). In the clinical setting dysfunction of the BBB or the BCB have to be differentiated, but dysfunctions of both, the BBB and BCB, may prevail in parallel in one individual patient. BBB breakdown may be demonstrated by MR imaging when fluid accumulation indicated local brain edema, which is not a highly sensitive finding in (meningo-) encephalitis (compared with Osborn et al., 2010), or by tracer imaging, rarely applicable in clinical approaches. BBB dysfunction cannot be diagnosed by CSF investigation. In general, the interpretation of CSF findings requires sophisticated expertise as from many possible influences CSF protein composition may change (see Wildemann et al., 2010). Recent findings demonstrated that neuroinflammation may often begin at the meninges (Bartholomaus et al., 2009; Ransohoff, 2009), this aspect vice versa was matching the clinical experience of an outstanding role of CSF investigation for differential diagnosis of neuroinflammatory disorders. 74 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 Table 4 Preliminary criteria for etiopathogenetic subgrouping of schizophrenias. 1. Genetic schizophrenia (gS): single gene trait model with strong weight for the gene defect, accordingly assessed in the individual case, and/or shown to be of considerable weight in population-based studies. 2. Neurodevelopmental schizophrenia (nS): early neurodevelopmental deficit or aberration, clearly assessed in the individual case and confirmed in population-based approaches to increase the risk for schizophrenia. Such may not be exclusive for schizophrenic syndromes, may for example include affective syndromes, or autism, which may precede or parallel schizophrenic syndrome. However, the aberrant neurodevelopment should be of considerable weight to induce in a major share of cases (specific) schizophrenic syndromes. 3. ME schizophrenias (meS): a LLNI process assessed by appropriate methods (CSF, neuroimaging, other laboratory methods) during critical disease stages. LLNI (strength and type) may vary, e.g. in between prodromal stage and other, more or less process-active, stages. 4. Primary nS with secondary meS: a special case when an early hit onto the immune system or onto basic metabolic cellular functions induced or considerably contributed to a late onset LLNI process. 5. Other types possibly to be differentiated with increasing knowledge. to a known CNS infection or CNS-specific autoimmune disease. Typical cases of primary meS would resemble prodromal meningoencephalitis or prodromal limbic encephalitis (LE). One might diagnose secondary meS when CNS involvement is secondary to systemic autoimmune or inflammatory disease (when assessed according to established rules), and the patient presents schizophrenic syndrome. A third category may be termed reactivated meS, in which an early infection was later reactivated during a critical disease phase such as the onset or an acute relapse. Long dormancy phases in infections (i.e., latency periods) are now well-documented, and therefore, such a separate category might be justified. A fourth category, called molS, could be considered in which the molecular pathomechanisms secondary to the infections of the CNS are involved and the molecular change is of sufficient strength to induce schizophrenic symptoms; however, this classification is still a speculative one. The question arises again of whether a border between LLNI and molS should be defined. In sum, there is broad evidence that beyond classical CNS inflammation (i.e., encephalitis), both with and without meningitis, a lower degree neuroinflammation, here termed LLNI, may prevail and be associated with (neuro-)psychiatric disease. LLNI represents the core pathogenetic aspect in a subgroup of psychiatric disorders, and LLNI, like classical neuroinflammation, is associated with a range of possible syndromes. In other words, the role of LLNI is nonspecific under an etiological view and heterogeneous under a categorical diagnostic view, and LLNI includes subgroups of schizophrenia (meS). The meS subgroup could alternatively be termed ‘organic’ or ‘symptomatic’. A definition of ME depends on the diagnostic armature employed to assess LLNI situations in vivo, i.e., the diagnostic sensitivity provided by the available methods. With advanced CSF analysis, we found that approximately 40% of therapy-resistant schizophrenia cases had some CSF pathology, which probably or at least possibly indicated LLNI (Bechter et al., 2010a). When using new methods such as microglia imaging or new antibody tests (as with LE, see below) to assess LLNI situations, the meS subgroup might be considerably enlarged. CSF investigation seems to be of utmost importance in assessing LLNI, and while neuroimaging and blood investigations play more supportive and indirect roles, they can nevertheless provide signals of CNS inflammation and, under certain circumstances, can be more sensitive in detecting inflammation than CSF investigation (for example in LE, where CNS-specific autoantibodies seem to be systemically produced). Differentiating a meS subgroup as its own category may be justified, the size of the meS subgroup might be considerable. 3. Human Borna disease virus (BDV) infection as an ME model: a slowly emerging research area Borna disease virus (BDV) infection was proposed as one etiology in the ME scenario that could be relevant for a maximum of 3% of cases in our psychiatric hospital sample (Bechter, 2001). The agerelated variance in pathogenicity (cases resulting in disease per cases of infection), a poorly explained but well-established phenomenon in infectious diseases, was discussed with regard to an ME subgroup of schizophrenia. The candidate was mumps virus infection (and, with preliminary data, also BDV infection), and the ages at which high pathogenicity (including mumps encephalitis) was observed matched with the age at which schizophrenia onset was likely; thus, it was hypothesized that there was a causal relationship. Additionally, in large epidemiological studies, the mumps virus has been shown to be a risk factor for psychoses, including schizophrenia (Dalman et al., 2008). BDV research provided a couple of models for the ME hypothesis. BDV is a strongly neurotropic virus. Natural BD has been diagnosed over centuries, and BD is the most frequently occurring meningoencephalitis in horses and sheep in Middle Europe (see reviews in Carbone, 2002). Outcomes of BDV infection depend on the strain of the virus, the patient's immune system and circumstantial factors such as the route of infection, among others (Herzog et al., 1997). BDV infection can trigger autoimmunity (Rott et al., 1993) or can cause behavioral abnormalities in primates (Sprankel et al., 1978); such findings resemble those found in human BDV research (compared with Bechter, 1998, 2001). Accumulated findings suggest that human BDV infection may be causally related to some neurological and psychiatric syndromes, including schizophrenia (Bechter, 1998, Herzog et al., 1997, Richt et al., 1997b). However, as research on human BD presented a challenge, the field began to lose interest (Wood and Bloor, 2006) and the widely differing findings in human BD research have raised major concerns (Schwemmle, 2001, Schwemmle et al., 1999), and controversy has evolved within the field (Bechter, 2001, Bode and Ludwig, 2003). A meta-analysis showed that the putative human BDV sequences identified mainly in blood samples may have been artifacts of laboratory contamination (Durrwald et al., 2007), which has been previously suggested (Schwemmle et al., 1999) and is consistent with previous negative findings (Richt et al., 1997a). Research on natural BD in animals detected rather clear epidemiological patterns, with regional genetic clustering of BDV strains that pointed to the existence of to-date unknown endemic reservoir host populations (Durrwald et al., 2006). It is important to note that a conclusive diagnosis of encephalitis has to be based on a complex clinical approach, which usually requires the inclusion of a CSF investigation, which is the gold standard. However, the sensitivity even of CSF investigations is limited. According to the results of the large California encephalitis project, even in the acute phases of meningoencephalitis, a disease stage of high diagnostic sensitivity, the specific etiology of the disease was only determined in fewer than 50% of cases (Glaser et al., 2006). Concerning the question of human BD, it is possible that a careful meta-analysis of blood investigations (Arias et al., 2012) can give some guidance to BD epidemiology, but this first requires a critical discussion of the laboratory methods used in the studies. It also seems highly questionable whether the fact and fantasy surrounding human BDV infections (Lipkin et al., 2011) can be resolved when the strict diagnostic rules established through substantial experience with human CNS infectious diseases in general were neither applied nor carefully considered in such evaluations. The recent findings of a multicenter study in the US of exclusive blood investigations of a K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 rather small number of subjects (Hornig et al., 2012) match with our longstanding findings and with the findings about BDV infection in animals but seem to provide little in the way of improved diagnostic sensitivity, which probably cannot be achieved through blood investigations alone. The newly developed ELISA should be validated in large epidemiological studies, including in animal populations, as was performed and used in our studies (compared with Herzog and Rott, 1980, Herzog et al., 2008). [It may be of notice that for validation of the newly introduced ELISA the sera from our well validated collection were used (compared with Hornig et al., 2012).] 3.1. Our research on human BD Applying strict diagnostic criteria, we investigated more than 20,000 patients over more than 25 years; this research involved more than 2000 MRIs and approximately 500 CSF investigations that were analyzed using various methods including PCR, and attempts were made to isolate a virus from CSF and post-mortem brains (Bechter, 1998, Herzog et al., 1997, Lieb et al., 1997, Richt et al., 1997b, Rott et al., 1991). Based on our evaluations and on clinical findings, we assumed that both neurological and psychiatric syndromes could be caused by BDV infection (Bechter, 1998, 2001). The specificity of the human BDV antibodies as assayed was confirmed through the recognition of linear epitopes of BDV proteins (Billich et al., 2002). In some psychiatric cases with previous BDV infection, which was evidenced by the presence of serum antibodies, CNS immunopathology was detected through CSF investigation (Oleszak et al., 2007). Nevertheless, in only a very few psychiatric patients BDV antibodies were produced within the intrathecal spaces, as observed through an increased antibody index (AI) (Bechter et al., 1995, 2010a). Increased AI (CSF/serum) is now considered to be a sensitive and valid indicator of specific types of encephalitis, usually found in the chronic or post-acute phases of infections (compared to Reiber and Peter, 2001, Wildemann et al., 2010). In some cases, AI may persist for a long time, a phenomenon that is not yet fully understood; however, a recent study on EBV infections with neurological diseases raised the possibility that co-infections might be responsible (Kleines et al., 2011). The validity of our diagnostic approach was further confirmed by comparison of blood, CSF and brains from naturally infected horses suffering from acute BD (Herzog et al., 2008). Nevertheless, the overall relevance of BDV infection for human psychiatric disorders may be overestimated; in our hospital population, it might represent a maximum of 3% of cases, but when using strict diagnostic criteria, we found only a few cases fulfilling the probable criteria of BDVassociated ME (see Bechter, 1998, 2010a, and unpublished), and none were definite. 3.2. Emerging pathomechanisms relevant in BDV infections Persistent BDV infection, virus reactivation, relapses or chronic courses of defects have been observed in animals. Many downstream pathomechanisms have been studied in BDV infection, including the roles of entry factors, reactive oxygen species, neurodegeneration, regeneration, neuroprotection, virus toxicity, inflammatory toxicity, inflammation-induced neurotransmitter alterations, neurotransmitter abnormalities induced by persistent noncytolytic infection, infectioninduced cellular dysfunctions including dopamine and serotonin dysfunction, among others (for details see Clemente et al., 2010, Daito et al., 2011a, 2011b, Dietz et al., 2004, Herden et al., 2000, 2005, Herzog et al., 1991, Kamitani et al., 2003, Koster-Patzlaff et al., 2007, Ovanesov et al., 2008, Planz et al., 2002, Pletnikov et al., 2002, 2008, Schwemmle and Heimrich, 2011, Werner-Keiss et al., 2008). Another pathomechanism was the upregulation of MHC class I-dependent targets for CD8 T-cells on neurons upon BDV infection and the production of cytokines in neurons, which both acted directly upon CNS inflammation (Chevalier et al., 2011). BDV sequences are integrated into 75 the human genome (Belyi et al., 2010, Horie et al., 2010), these findings raised new questions on the pathomechanisms possibly involved (Feschotte, 2010). The involvement of endogenous viruses in multiple sclerosis and schizophrenia has been specifically discussed for HERV-Ws (Perron and Lang, 2010, Perron et al., 2008). In sum, human BDV infection may be relevant for neuropsychiatric diseases, but only for a maximum of approximately 3% of hospitalized psychiatric patients, i.e., in less than half of the patients presenting BDV-specific antibodies. Human BDV infection may include a subgroup of schizophrenia cases, but only rare single cases fulfill the strict criteria for probable (low level or chronic) BDV ME and none yet appears to be proven. 4. Where we stand in the search for the causes of schizophrenia Presently, the neurodevelopmental hypothesis is generally preferred over the vulnerability stress hypothesis, but the former does leave some important questions unanswered (Owen et al., 2011). Puristic genetic hypotheses have waned in recent years with the release of results from genome-wide association studies: though an important role for genes in schizophrenia was corroborated, single genes appear to have a low impact, and while a number of confirmed risk genes were related to immune and basic cellular functions, only some were related to neurotransmitter abnormalities, contrary to previous speculations. In another important area of research, the brain imaging field, a neurodevelopmental versus a neurodegenerative model was heavily debated. Such major lines of research will be discussed here with respect to the ME hypothesis, with a focus on neuroimaging findings (see also Table 5). The 6th Symposium for the Search for the Causes of Schizophrenia in Sao Paolo, Brazil in 2009 came to the following conclusions (Kirkbride and Scoriels, 2009): variation in the incidence of schizophrenia exists; the risk for individuals depends on the type of environment they are exposed to; gene–environment-interactions may hold the key to revealing etiological pathways; the genetic load could be confirmed, but the outcome is characterized by largely unexplained variants; more evidence for a neurodevelopmental compared to a neurodegenerative hypothesis may exist; there was insufficient evidence to reject the possibility that psychotic symptoms are continuously distributed in the general population; and finally, the need for biomarkers should be stressed. In addition, reviews of genetic findings have come to the following conclusions: the underlying pathophysiology of schizophrenia remains largely unknown, there is some support for the dopamine hypothesis, and in agreement with others, a cluster of genome-wide significant snips lie in the MHC region, implicating the immunological system in the pathogenesis of schizophrenia (Stefansson et al., 2009). Another review (Bondy, 2011) has stressed the promising findings from genome-wide association studies, which have not met initial expectations of identifying a ‘susceptibility gene’. Incorporating new approaches such as epigenetic mechanisms or gene–environmentinteractions was proposed for future studies. Another view was that genes might be involved in entirely unexpected disease pathways, e.g., by copy number variance or deletions; however, such pathways were expected to lead to a spectrum of neuropsychiatric impairments rather than to a specific schizophrenic syndrome because the genetic overlap between diagnostic categories was strong (Rujescu, 2011). 4.1. Brain imaging According to repeated and refined analyses of imaging results, it seems rather clear that during a period of 10 years following the first episode of schizophrenia, a progressive brain change occurs that is characterized by a significant decrease in multiple gray and white matter regions and a corresponding increase of cerebrospinal fluid spaces, which is mainly associated with cognitive impairment 76 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 Table 5 Major characteristics of schizophrenia: present views. Overall view • Variation in the incidence risk depending on the type of environment • Genetic load important • Gene–environment-interactions may hold the key • Outcome characterized by largely unexplained variants • More evidence for a neurodevelopmental as compared to a neurodegenerative hypothesis • The underlying pathophysiology remains unknown • Some support for the dopamine hypothesis Genetics • Cluster of genome wide significant snips lies in the MHC region implicating the immunological system • Recent results not meeting initial expectations to identify a ‘susceptibility gene’ • Epigenetic mechanisms possibly involved • Gene–environment-interaction important • Not a specific schizophrenic syndrome related to genetic load Neuroimaging • Progressive brain change after first episode with significant decrease in multiple gray matter regions and multiple white matter regions and a corresponding increase of cerebrospinal fluid spaces • In the first ten years diagnostic classification changing • Widely distributed gray matter abnormalities early in illness likely due to neuropil elimination, progressive over the initial years of illness • From diffusion tensor imaging: a disorder of neural integration arising from white matter abnormalities • Frontotemporal connectivity abnormal • But: schizophrenia not localized in the brain! • Early and late hit • In part neurodevelopmental or neurodegenerative, disease process varying over time • Special rare findings: seemingly only during acute psychotic presumably process-active stages, one may observe slight brain volume increases or microglial activation Post-mortem • No evidence for classical inflammation but widespread pathological alterations, findings compatible with LLNI subgroups (results not outlined in detail here; compare Bechter and Bogerts, 2007; Steiner et al., 2011) (Andreasen et al., 2011, Fusar-Poli et al., 2011a, 2011b, Sun et al., 2009). The brain atrophy seems to be related to a closer packing of neurons and also to a decreased number of neurons (Smiley et al., 2011), which is influenced by different types of medication and the severity of the symptoms (van Haren et al., 2011). Identifying the types of medications that may influence the brain was complex, and outcomes may differ between different compounds, e.g., some may affect neuropeptide Y and corticotrophin (Nikisch et al., 2011), while others may directly interact with inflammation (Meyer et al., 2009). Furthermore, in the first ten years, the diagnostic classification changes, and it seems that a final diagnosis of schizophrenia can be linked to more severe or more active disease processes (Bromet et al., 2011). In the preface to a recent authoritative book on Neuroimaging in Psychiatric Disorders, the editors M. Shenton and B. Turetsky stated that the imaging field has provided a major in vivo contribution to a better understanding of schizophrenia and has even supported neuropathological approaches. This field has come to the following conclusive statements (Shenton and Turetsky, 2011): schizophrenia is associated with widely distributed functional brain abnormalities; widely distributed gray matter abnormalities are observable early on in illness and, ostensibly, premorbidly; gray matter reductions are likely due to neuropil elimination as opposed to neuron death; gray matter abnormalities are likely progressive, at least over the initial years of the illness; from the diffusion tensor imaging studies, schizophrenia appears to be a disorder of neural integration arising from white matter abnormalities, in part resulting from irregular myelin; in terms of functional aspects of the brain, frontotemporal connectivity specifically was found to be abnormal. However, it was difficult to interpret all of these findings, and an ‘extremely speculative hypothesis’ was proposed regarding the underlying mechanisms of schizophrenia; the hypothesis specified that, in any case, a late developmental trigger of schizophrenia has to be assumed (Whitford and Shenton, 2011). This conclusion was in agreement with the commentary of Nancy Andreasen published in the same book (Andreasen, 2011): the functional and structural brain abnormalities in schizophrenia have not been localized yet, nor has the timing of the changes been observed with sufficient clarity. The explanations for the imaging abnormalities found in schizophrenia have had to be shifted from the previous ‘early’ neurodevelopmental hypothesis to a late neurodevelopmental hypothesis, i.e., a late hit hypothesis, because the time trajectories seemed in part to be neurodevelopmental or neurodegenerative events with blurred boundaries. These recent conclusions match with previous claims that early and late neurodevelopmental changes may be observable (Pantelis et al., 2007). It should be noted here that, from the research on the course of clinical symptoms, it appears that the disease process itself may vary over time, from prodromal or basic stages to more process active stages (compared with Klosterkotter et al., 1989, Klosterkötter, 2011). With regard to the ME hypothesis, assuming that LLNI is relevant in diagnostically overlapping neuropsychiatric syndromes, findings about mood disorders may also be interesting to look at: as in the schizophrenia research field, the neuroimaging data in this field have not been consistent, but a set of brain regions has been described as critical to normal and abnormal mood regulation (Holtzheimer and Mayberg, 2011). Another interesting case was two definite developmental disorders and their manifestations in neuroimaging: complex changes in the brain's structure and functioning over time, including surprising local volume increases, have been found, and some abnormal developmental trajectories have rather clearly been assessed (Herrington and Schultz, 2011, Minshew, 2011, Santos and Meyer-Lindenberg, 2011). In comparison, the abnormalities of developmental trajectories in schizophrenia seem only to be partly categorized as aberrant neurodevelopmental trajectories or seem to be either rather late or at least not necessarily early (see also White et al., 2003, 2009, 2010, 2011a, 2011b). A rare study using short-term repeated imaging during acute psychotic phases showed brain volume increases that were normalized a few weeks later, cautiously interpreted as being due to slight brain swelling from inflammation during short-lived acute phases (Garver et al., 2008). Other rare studies have found microglial activation in the brain tissue of patients who died from suicide in acute psychotic phases (Steiner et al., 2008), and microglial activation has also been found during acute psychosis using PET in vivo (Doorduin et al., 2009). Microglia are sensors of pathological events in the CNS; they K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 are a parameter in CNS inflammatory responses (Kreutzberg, 1996, Raivich et al., 1999) and work protectively or destructively (Glezer et al., 2007, Saijo and Glass, 2011). Memory T cells persisting in the brain following an MCMV infection can induce long-term microglial activation via interferon-gamma (Mutnal et al., 2011). Microglia interact with other cellular systems and with volume transmission mode (see below). In explaining abnormalities of brain structure and function over time, one must consider a complex array of pathogenetic factors, including independent pre-existing and interacting factors. The need for this consideration can be illustrated through the example of cortical thickness, which is often investigated in schizophrenia. It appears that cortical thickness is in part genetically determined (Kochunov et al., 2011) and is influenced by medication (van Haren et al., 2011) and social stress, such as adverse childhood experiences (Benedetti et al., 2011). However, cortical thinning could clearly be a consequence of LLNI, trauma or various types of toxicity. Interestingly, in the schizophrenia subgroup with adverse childhood experiences, cortical thickness seems to be normal, whereas in a non-schizophrenic group, cortical thickness appeared to increase as a result of adverse childhood experiences; the interpretation was that the combined effect of cortical thinning from schizophrenia and cortical thickening from adverse childhood experiences resulted in a cortex that appeared to be normal with respect to the aforementioned schizophrenia subgroup (Benedetti et al., 2011). In sum, abnormalities and changes in brain structure and volume are frequently observed in the course of schizophrenia over time, from prodrome to the acute, chronic, and relapsing phases. These changes used to be referred to as neurodevelopmental, but during the last years, neurodegenerative aspects have increasingly been assessed. A consistently abnormal developmental trajectory related to one early hit has not been safely demonstrated. Consensus reports now suggest that the story is more complicated and that a so-called ‘late hit’ seems to be a minimal requirement in explaining the structural brain changes occurring during the early years of disease. The changes and dysfunctions over time possibly involve the whole brain, with no clear local preferences. Similar conclusions have been reached in the histopathology field (Steiner et al., 2011), not reviewed here. Disconnectivity, atrophy and complex structural alterations accompanied by dysfunction, or specific rare findings such as microglia activation and CSF pathology (see above) have yet to be explained. Apparently, there are many interacting factors and pathological aspects, including genetic and non-genetic factors, that influence the neurodevelopmental phenotype. When considering LLNI as a possible factor, it should be recognized that neurodegeneration is a typical consequence of neuroinflammation, but overall outcome depends on many interacting downstream factors, including neuroprotection, repair capacity, pre-existing factors such as stressful experiences, and medication, as well as factors that remain unknown. Present neuroimaging methods are rather sensitive to detect the indirect consequences of CNS inflammation, such as atrophy or structural abnormalities, and new imaging methods with high magnetic field strengths may further improve detection sensitivity and differential diagnosis (Bustillo et al., 2010). However, neuroimaging methods are often not sensitive enough to directly detect CNS inflammation, and they seem to be insensitive to LLNI situations. Therefore, a more detailed analysis of the diagnostic possibilities and limitations in supposed LLNI situations is of interest. 5. Diagnosis of CNS inflammatory disorders and LLNI The limitations on the detection of LLNI with currently available diagnostic methods may be examined in light of a look into classical inflammatory CNS disorders. The established diagnostic methods are neuroimaging and CSF investigation. 77 5.1. Neuroimaging Direct signs of CNS inflammation include water accumulation (swelling), blood exudation and local tissue destruction, especially visible in an MRI; frequent indirect signs represent meningeal enhancement (in the case of accompanying meningitis), and atrophy may become visible in the post-acute phase (Osborn et al., 2010). Both focal and distributed atrophy may develop over time in chronic neuroinflammation, the best-studied example of which was multiple sclerosis (MS) (Calabrese et al., 2009, Grassiot et al., 2009). Interestingly, the contributions of cortical gray matter atrophy and ventricle enlargement underlying neuropsychological impairment appear at least in part to be independent in MS (Tekok-Kilic et al., 2007). However, there are differences among different types of CNS inflammatory disorders. CNS involvement in generalized autoimmune disorders such as neurodermitis or systemic lupus erythematodes is generally difficult to assess (Bertsias and Boumpas, 2010). The type of atrophy in MS differs from that in generalized autoimmune disorders, though there is some overlap with regard to hemispheric white matter lesions (Coban et al., 1999). In an experimental autoimmune encephalitis (EAE) model, cortical atrophy correlated with disease duration when cerebellar white matter lesions were detected at an early time point, demonstrating that myelin-specific autoimmune responses can lead to brain atrophy in an otherwise normal CNS (MacKenzieGraham et al., 2006). However, local atrophies (for example, of the thalamus) may be observed in MS for unknown reasons (Houtchens et al., 2007), and atrophic process in MS can improve with stem cell transplantation (Rocca et al., 2007). 5.2. Developing limbic encephalitis Another example of previously often undetected neuroinflammation is limbic encephalitis (LE): prodromal stages, typically of several weeks, are associated with a variety of psychiatric syndromes. The diagnosis of LE is established with the onset of neurological hard signs or through findings from CSF or brain imaging and to a large extent by detection of CNS-specific autoantibodies. Slight brain swelling or minor hyperintensities may be observed early in neuroimaging and probably represent minor signs of encephalitis, but often, the neuroimaging results are normal. In some cases, hyperintensities observed in MRIs, which are a direct sign of neuroinflammation, have persisted for months or years, although progressive temporomesial atrophy usually develops without direct signs of inflammation (Urbach et al., 2006). Early LE ideally follows the scenario assumed in the ME hypothesis because various psychiatric syndromes are associated with an apparently emerging LLNI process (compared with Dalmau et al., 2011, Irani et al., 2010, Irani and Vincent, 2011, Pruss et al., 2010, Vincent et al., 2004, 2011). The short time of transition from the prodrome to the classical encephalitis stage strongly suggests that the psychiatric symptom spectra observed during preceding stages were associated with non-classical CNS inflammation, or with LLNI as proposed here as a term. Based on this evidence, the protagonists of LE research have speculated that the cases identified may represent only the tip of the iceberg (Vincent et al., 2011). Indeed, an increasing number of cases with prominent psychiatric symptoms were identified (De Nayer et al., 2009, Graus et al., 2010, Zandi et al., 2011); some showed symptoms such as epilepsy combined with psychiatric symptoms (Bien et al., 2007). The LE story again confirms the unspecificity rule of ‘hits’ to the brain; in the cases of low-level and classical encephalitis, the hit is associated with a broad spectrum of psychiatric symptoms that may change over time, and one individual case might pass through several nosological categories of psychiatric disorder during short time periods. It should be noted that the new laboratory methods to detect CNS autoantibodies have mainly improved the diagnosis of LE, whereas neuroimaging has often been inconclusive. 78 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 5.3. LLNI in schizophrenia Through use of advanced CSF investigation methods, we found approximately 40% of cases of therapy-resistant schizophrenia to be classified as possible or probable LLNI (or meS) (Bechter et al., 2010a), further supported and enlarged by cases demonstrating activated CSF cells by normal cell counts (Maxeiner et al., 2009). CSF abnormalities in schizophrenia have also been reported also by others (Holmes et al., 2006, Huang et al., 2006, 2007, Stanta et al., 2010), and all of these findings are consistent with ME hypothesis in general. Incomplete LE cases have also been suspected (see above), and indeed, in a preliminary study, CNS-specific autoantibodies were found in approximately 10% of cases with acute schizophrenia (Steiner et al., 2011). Findings about the schizophrenia subgroup having a previous HSV-1 infection coincide with the assumption of an LLNI process: the local gray matter differences were attributable to neither medication, chronicity, nor comorbid substance use (Prasad et al., 2007), and this finding was supported through the long-term observation of cases (Prasad et al., 2011). Similar approaches have subgrouped schizophrenia cases according to immune activation phenotype (Garver et al., 2003), which would be similarly consistent with the LLNI concept. Nevertheless, more specific and additional methods of investigation are needed for many reasons, not the least of which is the likely broad panel of infectious agents involved in schizophrenia, as has been suggested by epidemiological studies. In sum, the present methods of diagnosing CNS inflammation or classical encephalitis are of rather limited sensitivity, whereas the indirect signs of neuroinflammation, such as meningeal enhancement and atrophy, the latter of which develops with some time delay, are sensitively detected. It is nearly impossible to detect diffuse low-level CNS inflammation with present imaging methods, although such LLNI situations have been documented to prevail over considerable time periods in experiments and in human disease, e.g., during the prodromal stages of LE. Indirect signs of LLNI in schizophrenia may be mild atrophy, minor local hyperintensities, or local swelling over short time periods. In the meS subgroup (Table 6), several or many infectious agents seem to be involved, and differing types of LLNI may evolve as a result. Specific types of encephalitis may differ from others in important ways; e.g., HIV encephalitis (HIVe) is characterized by a continually high virus load within the brain. This feature strongly differs from those found in most other CNS infections, but even in HIVe, the search is on for more sensitive methods (Bucy et al., 2011). The clinical symptomatology involved in both classical meningoencephalitis and LLNI situations may generally include a spectrum of psychiatric symptoms or syndromes, and with increasing severity of the inflammatory process, more and more neurological hard signs or ‘organic’ psychiatric symptoms may evolve. The symptom spectra in CNS inflammation are thought to be influenced by pre-existing independent factors and liabilities, not the least of which are genetic. The rare studies performed on schizophrenias using sensitive methods such as microglia imaging or CSF investigations produced results supporting the view that LLNI may be underdiagnosed in psychiatric disorders in general and in schizophrenia specifically; this was also strikingly evidenced by recently detected LE-associated psychiatric disorders. CSF investigation appears to represent the gold standard for research into the meS subgroup. This is not contradicted in principle by recent experiences with LE because a strict diagnosis of CNS inflammation is also difficult in LE; however, methods for detection of the CNS-specific autoantibodies in the blood are sensitive in LE, which is highly suggestive of an inflammatory involvement of the brain. Nevertheless, any safe diagnosis of CNS inflammation and especially of LLNI requires a broad clinical approach. 6. Neurodevelopmental, genetic, ME and other schizophrenias? Neurodevelopmental schizophrenia was conceptualized as an early, usually prenatal, hit during neurodevelopment. In experiments, the timing rather than the type (infectious agent, immune response factors) of hit was important for disturbed neurodevelopment (Meyer et al., 2006). This finding resonates with the epidemiological evidence on pre- and perinatal hits in schizophrenia for factors such as hunger (St Clair et al., 2005, Susser and Lin, 1992), infections or birth complications (Brown, 2011a). Premorbid or inborn aberrations such as minor physical anomalies are more frequent in the schizophrenic population (Dean et al., 2006), and individuals with these abnormalities may constitute a subgroup with an increased risk of schizophrenia. Animal experiments have not covered the whole neurodevelopmental period, nor have studies of humans, but covering the whole developmental period is apparently necessary, as timing seems to be the major determinant of neurodevelopmental aberrations. Recent transcriptome analysis has enlarged the number of periods that should be considered: 7 periods of embryonic or fetal development, 5 periods from birth to adolescence, and 3 periods in adulthood (Kang et al., 2011). To define neurodevelopmental schizophrenia, study of these 15 different neurodevelopmental periods and their associated specific aberrations is required. The typical age of schizophrenia onset should match with the transcriptome-based categorization of young adulthood period, defined as 20 years ≤ age b 40 years. However, transition to psychosis might be a process-active stage (Klosterkotter et al., 1989) and thus not necessarily a neurodevelopmental process, and the strong claims about a ‘late hit’ (see above) present a challenge to Table 6 Major pathomechanisms and sites involved in LLNI. • Brain barriers: BBB, BCB, meningeal–CSF barrier (see also Table 3) • Endogenous CNS immunity: established by and involving probably all types of CNS cells, especially microglia and astrocytes, and even neurons, and humoral signaling (McGeer and McGeer, 1995). • Wiring transmission (WT): mainly represented by synaptic transmission, WT is balanced by VT (see Fuxe et al., 2010). • Volume transmission (VT): mainly mediated by extracellular CNS fluid (ECF), moved by the pulsatile brain throughout the extracellular CNS spaces, ECF being in exchange with CSF. VT model includes many different signaling molecules, e.g. cytokines, reactive oxidative species, gasses, and many others. Effects depend from the location where interaction takes place and what receptors are expressed at resident cells. • CSF cells: play a previously underestimated role for CNS functioning and CNS immunity in health and disease (Schwartz and Kipnis, 2011; Schwartz and Shechter, 2010). • CSF signaling: incompletely investigated, apparently involving signals from various sources, including the choroid plexus, hypothalamus, CSF cells, and others, and with links to ECF • Peripheral cerebrospinal fluid outflow pathway (PCOP): CSF is flowing out through cribriform plate and along all brain nerves and peripheral nerves into peripheral tissues (Cserr and Knopf, 1992). A possible interaction between CSF contents and nerves at peripheral sites and in addition with peripheral tissues was hypothesized to possibly explain unexplained findings in subgroups of major psychoses including schizophrenia, and in neuroinflammatory disorders in general (Bechter, 2011). Also CSF cell trafficking was demonstrated through the cribriform plate (Goldmann et al., 2006; Kaminski et al., 2012) and in preliminary study along peripheral nerves (Schmitt et al., 2011b). • Molecular mechanisms: molecular transport mechanisms play a major role for the exchange at the brain barriers (Redzic, 2011). • Anatomical distribution of pathologies in schizophrenia: a common pathogenetic link of the distributed dysfunction and abnormalities of CNS found in schizophrenia is open. VT (and CSF signaling) extending according to the PCOP hypothesis into periphery (see above) could explain distributed intraparenchymal and distributed cortical involvement and in addition involvement of peripheral parts of nerves (as for example shown for the olfactory system) and even the involvement of peripheral tissues like muscle lesions. Thus the LLNI scenario may prevail widely distributed anatomical sites in parallel and thus could in principle represent a common pathogenetic link for anatomically distributed but pathophysiological similar dysfunctions (Bechter, 2011). K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 the validity of the neurodevelopmental model for a majority of schizophrenia cases. 6.1. Early and late hits to the brain? What is meant by a ‘hit’? Does this term refer to the onset of a true neurodevelopmental aberration, or does ‘hit’ simply mean that some pathogenetic process seems to go on? In the literature, it seems that some event early in neurodevelopment was understood as an early hit, whereas in the case of the recently introduced term ‘late hit’, it is rather unclear what is meant; it seems to refer to something that has happened to the brain. Various types of insults to the brain could be considered, including an LLNI process, which could combine (or not) with early neurodevelopmental aberrations. Until now, the question has remained open as to whether an early hit in schizophrenia is definitely necessary to produce schizophrenia, whether such a hit represents just one possible risk factor, or whether an exclusive neurodevelopmental trajectory is in play. When applying strict diagnostic rules, neurodevelopmental schizophrenia should only be diagnosed when an early hit induced neurodevelopmental abnormalities and the early hit was clearly assessed in the individual case and placed into a defined developmental period with appropriate criteria regarding the timing of the associated developmental abnormalities. Such strict rules seem justified in comparison to several other defined neurodevelopmental diseases, including monoamine neurotransmitter diseases, autism spectrum disorders or intellectual disabilities (see below). Another possibility is that a late hit was directly related to an early hit, but the early hit was to the immune system (Gorczynski et al., 2011). This possibility would correspond to both the neurodevelopmental hypothesis and the ME hypothesis; however, the early hit would not induce the developmental aberrations of the CNS, and the late hit could well be related to an LLNI process. Such a scenario, however, does not match with the usual neurodevelopmental concept of schizophrenia, for which a specific developmental trajectory has been proposed, whereas inflammatory mechanisms mentioned as a possibility were not matching with the neurodevelopmental model as proposed (compared with Brown, 2011b). Late consequences from early hits could also result from epigenetic processes or from genes involved in the metabolic pathways of DNA biosynthesis that may take place at many time points during life and seem to represent common risk factors in schizophrenia, bipolar disorder and unipolar depressive disorder (Peerbooms et al., 2011). In such types of early hits to basic metabolic functions, the later consequence might be similar to that resulting from a hit to the immune system: an increased liability to a late-onset LLNI process. 6.2. Unspecificity and heterogeneity in neurodevelopmental disorders and in CNS inflammation Overall, present knowledge suggests that schizophrenia is a heterogeneous disorder (Brown and McGrath, 2011), and this matches with the unspecificity rule for any hit to the CNS, including infections or LLNI. This also holds true for genetic disorders, though some relative specificity is possible (compared with monoamine neurotransmitter disorders). Thus, in general, emerging ‘specific’ schizophrenic symptoms may be related to whether the brain systems involved are able to produce such symptoms, as occurs in neurological disorders, and may become prominent with the increasing severity of pathology and dysfunction of the brain systems involved. Some symptoms may appear as non-specific, while others may appear as rather specific or easily identified at the psychopathological level. The latter may primarily depend upon pre-existing genetic liabilities and only in part on the pathogenetic process itself, which may, however, be central to the disease (compare also 7). 79 In defined neurodevelopmental disorders, more than 100 different gene defects are identified as causing intellectual disability and an increased risk for several psychiatric and neurological disorders. The causal genes are often involved in fundamental cellular processes that are pivotal for normal brain development and function, whereas only a few genes are involved in synapse function, contrary to previously speculation (Najmabadi et al., 2011). In prospective studies on several developmental disorders, including autism, some differences between single gene defects have become apparent, but mainly broad overlap and similarities have been observed: findings of minor specificity were that in autism associated with generalized overgrowth more severe symptoms were associated with more overgrowth (Chawarska et al., 2011). The complexity of early growth patterns in disabled populations was considerable, showing different types of disturbed development that deviated from controls with typical development, and interestingly, overlap was particularly observed in bodily and social impairments but not as much in brain developmental trajectories. These recent insights are of special interest here because both autism and intellectual disabilities are associated with an increased risk for schizophrenia-type psychoses and other psychiatric disorders. 6.3. Early infections Others have recently presented splendid reviews on the role of early infections in schizophrenia (Brown, 2011c, Brown and McGrath, 2011, Fatemi et al., 2012). The risk attributed to three early infections (by Toxoplasma gondii, herpes simplex virus, and rubella virus) was calculated to explain 33% of schizophrenia cases in the population. However, when looking into the details of what is possibly the best-studied case of human intrauterine infection, the rubella endemic in New York, the story appears to be complex (compared with Brown et al., 2001): approximately one half of offspring exposed to rubella during pregnancy became psychiatrically diseased in later life, suffering from a broad spectrum of psychiatric disorders including schizophrenia. Thus, again in light of this case of intrauterine infection, the unspecificity rule with regard to psychiatric syndromes has clearly been confirmed. For postnatal infections, the unspecificity rule is also evident. The world influenza epidemic, which was often cited as an example of an intrauterine hit, was also complex; Menninger reported mainly adult cases of exposure and, to my knowledge, not one single intrauterine infection. The population surviving the influenza infection experienced a considerably increased prevalence of a spectrum of neuropsychiatric disorders, including depression and schizophrenia, shortly after the influenza infection (Menninger, 1994). 6.4. Complex disease scenario Apparently, cohorts with definite abnormal neurodevelopment (and abnormal systemic development) show an increased schizophrenia risk, but the aberrant neurodevelopmental trajectories in themselves do not lead, at least not in the majority of cases, specifically to schizophrenia. Nevertheless, a neurodevelopmental schizophrenia (nS) subgroup likely prevails. The single causal factor of aberrant neurodevelopment has an apparently strong influence on the development of schizophrenia, although the symptoms themselves may be shaped by interacting pathogenetic aspects. In this scenario, a late hit seems unnecessary or would have little weight in the development of the disease. However, when an independent factor with a strong weight such as a late hit in addition to an early hit is needed to induce the disease, this is representative of a two-factor model, and the disease should not simply be termed a neurodevelopmental disorder. Conversely, when an early infection does little or no harm if it is not reactivated later, the major pathogenetic event determining the disease may even be considered to be the late one, and the early hit would also constitute a necessary risk. At present, the research base needed to assume that there 80 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 are definite aberrant neurodevelopmental trajectories in schizophrenia in general is not sufficient, although a subgroup with aberrant neurodevelopment and an increased risk for schizophrenia can be assumed beyond doubt to prevail. In sum, LLNI situations may possibly be involved in a late hit model of schizophrenia. It is informative to consider the role of risk genes in autism and intellectual disabilities with rather clear neurodevelopmental trajectories, at least in the majority of cases (see below), in addition to other slowly emerging diseases such as autoimmune disorders: risk genes represent liability factors but seem to weigh heavily in only a minority of cases (see Fathman et al., 2005, Rioux and Abbas, 2005 and below). Longstanding experience with the prodromal phases of classical meningoencephalitis (compared with Bechter, 2001) and recent examples such as LE clearly suggest that undetected LLNI has the potential to explain the late hit in a schizophrenia subgroup. Such an assumption is corroborated as complex genetic traits that seem to be relevant in the majority of cases of schizophrenia, and this coincides with a gene– environment interaction scenario, which is often stressed as necessary. Nevertheless, a gene–environment interaction could also be involved in a neurodevelopmental subgroup. For the differential diagnosis of various schizophrenia subgroups, it was necessary to define the major pathogenetic constraints, particularly during the disease-active stages, that prevail during critical disease periods from prodrome to transition or during relapse in chronic stages, during which some LLNI process may go on nevertheless (see also the CSF findings cited above and the comparison to autoimmune disorders). 6.5. New old problems in schizophrenia research The present difficulties in schizophrenia research seem to not differ much from previous discussions about the neuropathology of schizophrenia (Harrison and GRoberts, 2000). In a critical foreword from Janice Stevens, three major concerns about the neurodevelopmental model were noted: 1. the question of pathological heterogeneity and its relationship to the clinical syndrome; 2. the controversy concerning the location of the pathological changes; and 3. the timing of pathology. 6.6. Neurotransmitter disorders Bearing in mind these concerns, the research into neighboring disorders of abnormal neurotransmission may first be considered: Parkinson's disease was the first disorder for which an underlying neurotransmitter abnormality was identified. Parkinson's disease, meanwhile, is not explained by dopamine abnormalities alone, which are part of a complex disease process with a yet unknown etiological background (Braak and Del Tredici, 2008). The dopamine hypothesis of schizophrenia is widely accepted, and dopamine appears to be undoubtedly important in schizophrenia (Carlsson et al., 2004, Rolls et al., 2008) and has been shown to be directly involved in prodrome (Fusar-Poli et al., 2011c). The interpretation of the antipsychotic occupancy of dopamine receptors in schizophrenia is moving towards a better understanding of antipsychotic drug development and therapeutic approaches (Nord and Farde, 2011) rather than towards an understanding of the whole disease process. Such a change in interpretation seems very similar to that experienced in Parkinson's disease. Looking into the details regarding the heterogeneous group of monoamine neurotransmitter disorders (Kurian et al., 2011) is further informative: a developmental delay was accompanied by a spectrum of neurological and neuropsychiatric symptoms often beginning in infancy or early childhood, although onset may occur at any age. Pterin, dopamine and serotonin metabolism disturbances can be primary, or secondary, or of unknown origin. The diagnosis is based on a detailed clinical history and physical examination, appropriate assessments of neurotransmitters in the CSF, genetic screening, and the exclusion of mimicking disorders. The etiology is often genetic, and a schizophrenia subgroup has not been described. This nevertheless indicates that single genetic causes of aberrant neurodevelopment can produce a complex syndromal pattern, that differing ages of onset can occur, although onset usually occurs early in infancy (as for autism spectrum disorders and mental disability), and that the clinical approach requires multilevel diagnostics. Interestingly, CSF investigation, but not blood investigation alone, may be able to demonstrate neurotransmitter imbalance (Kurian et al., 2011). 6.7. Timing of hits and diagnostic and therapeutic approaches In schizophrenia, early neurodevelopmental hits that have been identified include hunger and infections (compared with Brown, 2011c, Susser and Lin, 1992); late hits that have been identified include cannabis use (De Hert et al., 2011). Many infections, according to recent large epidemiological studies, are associated with an increased risk of various psychoses, including schizophrenia (Brown, 2011a, Brown et al., 2004, Buka et al., 2001, Dalman et al., 2008, Gattaz et al., 2004, Koponen et al., 2004, Niebuhr et al., 2008, Westergaard et al., 1999, Yolken and Torrey, 2008). In large studies from Denmark, both autoimmune diseases and late infections additively and considerably increased the risk of schizophrenia (Benros et al., 2011), whereas early infections did not or did so only a little (Nielsen et al., 2011). However, epidemiological studies are not the only type of study that can be informative; it could be that a number of agents are not yet known, as was clearly suggested, for example, by the results of the large California Encephalitis Project, in which slightly less than 50% of cases were clearly identified, even in acute meningoencephalitis (compared with Glaser et al., 2006). Therefore, it is also of interest to perform small studies searching for specific viruses, e.g., parvovirus B19, which was associated with both autoimmune disorders (Pugliese et al., 2007) and psychoses (Adamson et al., 2011, Hobbs, 2006), or for BDV (see above). Regarding the question of heterogeneous schizophrenias under a categorical view, critical phases during the course of the disease, or so-called process active stages (Klosterkotter et al., 1989), may be the key to a better understanding of the disease itself; it appears to be of particular interest that only a portion of the at-risk cases will ever transit from the prodromal disease stages to full-blown psychoses (Klosterkötter, 2011, Riecher-Rossler et al., 2009, Simon et al., 2011). Diagnostic approaches especially have to consider the timing of the diagnostic intervention with regard to the interpretation of the results, as can be seen with an extraordinarily well investigated neuroinflammatory disorder, multiple sclerosis (see below). In sum, there is increasing evidence that schizophrenia is a heterogeneous disorder. Therefore, appropriate research and terminology should be available for delineating in a balanced manner the respective weight of the contributing factors involved in what is likely to be a complex etiopathogenetic scenario of risk factors, hits and modulatory factors. Early hits in general increase the risk of disease, but this may not be easily detected through cross-sectional investigation in the clinic. Nevertheless, factors or syndromal constellations assessed carefully during critical disease stages such as the acute psychotic phase at the disease onset or during a relapse would carry heavy weight with regard to the underlying pathogenesis, at least for that specific stage. Whether early risk factors may be necessary or only possible risk factors remains very difficult to determine. Major pathogenetic factors may also combine and thus be of note for subgrouping. As long as there is no definite knowledge that a neurodevelopmental aberration is needed, patients with such a history should be not considered to constitute the only pathogenetic subgroup. Here, preliminary categories for subgrouping heterogeneous schizophrenias are formulated (see Table 4); however, no operational criteria are included, as they would need to be determined through a consensus procedure. The results of recent elegant studies (Clarke et al., 2011, Dickerson et al., 2012, Wang et al., 2011) point to such subgrouping. Thus it was tempting to probe the proposed K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 etiopathogenetic classification including a categorization of diagnostic strength by such datasets at the individual case level. 7. Gene–environment interaction and ME hypothesis A major point consistently stressed is that environmental factors should play a more important role in schizophrenia than previously assumed and that neither genes (Bondy, 2011), though significant, nor the dopamine hypothesis (Nieratschker et al., 2010) may fully explain the etiology of psychoses. It is clear that more research is needed (Insel, 2010). In an insightful and critical discussion about the neurodevelopmental hypothesis (Fatemi and Folsom, 2009), the role of a gene–environment interaction in the pathogenesis of schizophrenia was not doubted, and viral and bacterial infections were addressed as candidates. 7.1. Environmental factors in schizophrenia and in autoimmune disorders Among the candidates for environmental factors, one might differentiate between risk factors and contributing factors, both of which play a possibly variable role in an interactive pathogenetic scenario over time. To differentiate required factors (conditio sine qua non) from secondary contributing factors, it may be especially important to focus on the eventual transition from risk states to diseased states. A telling example of such a transition is the onset and course of autoimmune disorders (Fathman et al., 2005, Rioux and Abbas, 2005), most of which relate to complex genetic traits, as does schizophrenia. General autoimmune disorders require three factors — genes, the environment and the immune system — which may become relevant during the disease process at different time points and even repeatedly at different pathogenetic levels. Autoimmune disorders may be initiated by infections via a couple of pathways (Mills, 2011), and disease outcomes seem to be considerably shaped by (local) tissue responses (Matzinger and Kamala, 2011). Similarly, when trying to better understand the complex pathophysiology of schizophrenia with respect to the gene– environment interactions involved, the inclusion of such new immunological concepts seems necessary. Among the environmental factors identified in schizophrenia are urban living and migration, the responsible pathogenetic mechanisms are thought to represent social stress (van Os et al., 2010). Specific mechanisms have been identified (Lederbogen et al., 2011). However, stress factors may be easily overestimated, as shown in previous examples, such as gastroduodenal ulcers, and possibly in schizophrenia as well (Kirkbride and Scoriels, 2009). Conversely, infections causing chronic diseases have often been difficult to identify; prominent recent examples include helicobacter pylori infection causing gastroduodenal ulcers (compared with Marshall, 1990), papilloma virus infections causing cervical cancer (compared with zur Hausen, 2001), and, years ago, two important neuropsychiatric disorders: the viral etiology of poliomyelitis and the spirochetal etiology of general paresis and tabes dorsalis. The etiologies in both of these latter cases were difficult to prove and hardly accepted (Bechter and Hodgkiss, 1995). In retrospect, the major hurdle for causality inferences in such infections has always been the overall low pathogenicity (i.e., the number of diseased per the number of infected) within the population. With regard to schizophrenia, environmental factors resulting in exposure to pathogens in people who may not have developed antibodies or pathogen resistance factors (e.g., newly acquired pathogens are more harmful for immigrants not used to these pathogens) are another option (Fatemi and Folsom, 2009), although there may be some contribution of social stress as well. 7.2. Gene–environment interaction in psychiatric disorders in general A most intriguing model of a gene–environment interaction for psychiatric disorders, including schizophrenia, has been presented 81 by Uher (2009): the evolution-informed framework (EIF) hypothesis elegantly explains the interaction between environmental and genetic factors over time within populations. The critical point, that risk genes prevail despite a strong negative impact of the disease on the number of offspring produced, suggests that environmental factors must be evolutionary recent. Uher mentioned infectious agents only in a co-authored paper (Caspi et al., 2010). Here, it is proposed that infectious agents appear to be ideal candidates for evolutionarily recent environmental factors, but they remain difficult to identify (see above). It is intriguing to combine the EIF hypothesis with another informative epidemiological scenario described in a milestone observational study of wild sheep: a complex interrelationship between survival, number of offspring and adaptive immunity to changing environments was found. In short, fitness correlates of heritable variation in antibody responses were related to variance in reproduction over the long term, and self-reactive antibodies were associated with adult survival during harsh winters, depending on the variance of the pathogen load (Graham et al., 2010). Such a scenario of natural selection and autoimmune responsiveness related to genetic variation and changing environment would be coherent with the pathomechanisms suggested in an meS subgroup, and it could explain a considerable subgroup of schizophrenias overall because the two major findings, the continuous genetic risk level within the population and the late hit, could be explained with ME. In a categorical view, a major or ‘causal’ role for the suspected LLNI process may be attributed to the immune system because the immune system is mainly responsible for adaptation to the environment. With regard to an epidemiological scenario, exposure of the individual to the specific infectious agent may be considered to be most important and relevant if prevention of the infections were possible; however, it is problematic that ubiquitous infections with overall low pathogenicity are hardly preventable. Therefore, from a practical perspective, the immune system in such a scenario is of the greatest relevance. However, the positive and negative effects of such genes may come to be recognized only in relation to certain infectious agents prevailing within the population. This is illustrated, for example, by the major changes of immune reactiveness that have evolved with civilization; e.g., the eradication of helminths seems to have shifted TH-1–TH2-immune-responsiveness to fall under other mechanisms, thus explaining the increasing prevalence of autoimmune and allergic diseases, a scenario referred to as the hygiene hypothesis (Allen and Maizels, 2011). 7.3. Contributive environmental and developmental factors Beyond the primary pathogenetic factors discussed above, there may be secondary contributive environmental factors as well as completely independent developmental factors; the latter may be neither early neurodevelopmental in nature nor involved in neurodevelopment, but may rather be involved in general development, including immune system development. Epigenetic dysregulation by environmental factors has been discussed in detail for HERV-W expression in different tissues relevant in multiple sclerosis and schizophrenia and for related immune responses with regard to the molecular pathways involved in inflammation (compared with Perron and Lang, 2010). For HERV-W, a life-long scenario of a determinant interaction between infectious agents and HERV-W genes has been proposed to explain the actual development and cause of schizophrenia (Leboyer et al., 2011); this scenario would also perfectly match with the ME hypothesis. Overall, it appears plausible that a complex scenario involving an interaction between the immune system, genes and changing environments may be implicated in schizophrenia and in LLNI. Two especially critical aspects of the epidemiology of schizophrenia could nicely coincide with the gene–environment interaction and the meS subgroup as defined here: 1. The preferred age of schizophrenia onset, because it 82 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 matches with the age-related variance of pathogenicity (high pathogenicity during the preferred age of onset) of a number of infectious agents. Long-known modulating factors of schizophrenia onset, such as stress, sex and hormones, would also match because they also modulate immune-inflammatory responses. 2. The continuously high prevalence of schizophrenia risk genes in the population despite a strong disadvantage regarding progeny may interrelate with advantages for survival lent by the heritable variation of immune responses with varying pathogen loads (compared with the wild sheep study above). The burden of schizophrenia for humans and the disadvantage for the CNS may represent the negative side of the immune system's associated advantages for survival under changing environmental conditions. Infectious agents involved in such a scenario are expected to have low pathogenicity overall, as they are well-adapted to by the host and lead to harm in only a few of the infected, but to have the potential to becoming pathogenic under certain environmental circumstances (e.g., co-infections and others) and in response to age-related variations of the immune inflammatory response (see also Bechter, 2001). Immune inflammatory responsiveness is the major adaptive system that living organisms use to cope with the environment. Recent insights into CNS immunology suggest that the CNS is not spared from immunity (see fallen immune privilege); instead, the CNS seems better protected, and CNS-specific inflammatory mechanisms are more complex than immune responses in other tissues, thus increasing the general plausibility of the ME concept. Previously unprecedented interactions are just beginning to be understood to have surprisingly strong impacts on various disease outcomes, e.g., the influence of gut microbiota on inflammation, autoimmune disease and cancer (Tlaskalova-Hogenova et al., 2011). In sum, gene–environment interactions represent an important aspect in the etiopathogenesis of schizophrenia and could be involved in various subtypes of schizophrenia; it is highly plausible that they are involved in the proposed meS subtype. Pathogenetic processes involved in schizophrenia vary over time, and one early and one late hit were proposed; however, the specific characteristic pathomechanisms of these hits remain to be better defined. The longstanding disease process that occurs over years must be explained, not only in terms of the associated consequences for the function and structure of the CNS, as well as the associated epidemiological aspects of disease course, but also in terms of the consequences for the development of the gene pool within the population over time. The required factors in autoimmune diseases are genes, the environment and the immune system, and the environmental factors often seem to be infectious agents, although they have only been partially identified thus far. A rather similar scenario could pertain to the meS subgroup because ME could well explain even a long-term disease process, whether it is initiated from latency after early infection and reactivation or from a late infection that interacts with previous infections and other interacting factors that have become relevant over time. Additionally, ME and the immunegenetic background could coincide with population-related aspects of risk increases under changing environmental factors, with downstream consequences for the whole population, which may change direction over time. As the meS subgroup could elegantly explain several major aspects of schizophrenia and its epidemiology, such as the continuously high level of risk genes within the population despite the disadvantage with regard to the number of progeny, as well as the preferred age of onset, this subgroup could be of considerable size. The most specific findings from CSF studies would support such view, although CSF analysis is not yet considered to be sensitive enough with respect to other available methods (compared with Bechter et al., 2010a). A prevailing LLNI pathomechanism, the core aspect of the ME hypothesis, remains difficult to assess in vivo. A full understanding of disease pathomechanisms would also require a characterization of the respective immune response states of the organism over time. From this characterization, the complex interactions that occur over time could be better understood and categorized as either classical inflammation at some time points or as intermediate states of immune regulation or struggles with the environment based on the individuals immune system at other time points, a model which has been outlined in principle in computational immunology (compared with Cohen, 2007). 8. An emerging ME scenario Here, further pathomechanisms involved possibly in the meS scenario are more closely examined. It remained difficult to delineate the precise role of infectious agents identified as schizophrenia risk factors with regard to the pathogenetic mechanisms involved (Yolken et al., 2011). Some agents seem involved at the onset of disease (Bechter, 2001, Dickerson et al., 2007, Leweke et al., 2004), suggesting an initiating role; however, this remains to be proven. Furthermore, this verification must be conducted separately for each agent because each agent associates with specific aspects of this virus or retrovirus, e.g., endogenous retrovirus HERV-W (Perron et al., 2008) or herpes virus 6a which is directly involved in the immune-inflammatory response (Arbuckle et al., 2010). Such differences were demonstrated when comparing for example just three well investigated CNS infections (Bentivoglio et al., 2011). 8.1. Persistent infections Emerging knowledge about persistent, latent or dormant infections demonstrated that transitions between dormant and reactivated or partially active infected states may be more important for disease than previously assumed (Dworkin and Shah, 2010, Kerschensteiner et al., 2009). The complex scenario of persistent infections, involving both good and bad for the infected individual, was demonstrated in detail for the Helicobacter pylori infection (Arnold et al., 2011, Willyard, 2011). Interactions between different infections in one infected host were increasingly recognized, though they are not reviewed extensively here. Furthermore, unconventional, inherent determinants may precipitate a diseased state only when an infection was present (Cadwell et al., 2010). It is worth noting that mechanisms were subject to considerable change over time (Bentivoglio et al., 2011). While persistent viruses and bacteria can exist over the long term in metabolic inactive states and thus survive without growth, dormant cells may sense when conditions improve and reinitiate growth (Dworkin and Shah, 2010). In a surprising clinical example, a dormant virus was unprecedentedly involved in peripheral immune response, first presumed to be an adverse drug event, but when studied in more detail, it appeared to be related to a reactivated EBV infection triggered by the drug. Thus, transformed B-lymphocytes induced the cutaneous and vascular symptoms of DRESS (drug reaction with eosinophilia and systemic symptoms) (Picard et al., 2010). 8.2. CNS endogenous immunity Many new aspects of CNS-specific endogenous immunity have been explored (Bentivoglio et al., 2011, McGeer and McGeer, 1995) and may plausibly be involved in LLNI. For instance, synaptic dysfunction was likely involved in meS, LLNI may be either primary or secondary effect from infection, autoimmunity or other insults, as disturbed neuronal transmission plausibly represents the functional extreme of any CNS pathology. However, synaptic dysfunction may often not demonstrate the primary disease process, as found even for the majority of neurotransmitter disorders identified to date (see above). A seemingly simple fact should also be considered; that is, neurons represent only a minority of CNS cells and need continuous support from glia, an aspect which was underestimated in CNS research including schizophrenia research (Nave, 2010). Additionally, the role of microglia, which is easily activated, is only beginning to be understood in more detail. This will not be further discussed herein. K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 8.3. Volume transmission Another basic mechanism rarely discussed in context with schizophrenia but of apparent importance for normal CNS functioning and, thus, likely for disease, is the volume transmission (VT) mode concept. The VT mode balances the so-called wiring transmission (WT), the latter represented mainly by synaptic transmission (Agnati et al., 1995). Pathological alterations of the VT mode can be assumed to be involved in the LLNI processes because CNS extracellular fluids provide general signaling functions throughout the CNS (Fuxe et al., 2010). Furthermore, VT links to CSF signaling (in two directions), thus the functional relevance within the CNS parenchyma is clearly demonstrated (Fuxe et al., 2010, Lehtinen et al., 2011). The role of the extracellular fluid space in CNS infections was illustrated in Toxoplasma infection where a rapidly developing kinesis-associated system of reticular fibers plays an important role for the movement of infiltrating immune cells to defeat toxoplasma (Wilson et al., 2009). Toxoplasma was one specific risk factor in schizophrenia (Yolken and Torrey, 2008). However, it has also been determined that sterile inflammation can be activated within the brain by non-specific sensing mechanisms (Chen and Nunez, 2010, Mills, 2011). 8.4. Downstream mechanisms Pathomechanisms may primarily or secondarily be involved in immune-inflammatory responses within the CNS in the form of alterations of neurogenesis (Belarbi et al., 2011, Ekdahl et al., 2009), oxidative stress (Trushina and McMurray, 2007), antioxidant capacity (Gawryluk et al., 2011), chronic neuroinflammation and neurodegeneration (Lynch and Mills, 2012), neuroprotection (Ehrenreich et al., 2008, Hellweg et al., 1998), autoimmunity (Jefferies et al., 2011), immune neural glial interactions (Lynch and Mills, 2012), CSF cells (Schwartz and Kipnis, 2011), infection- or inflammation-associated neurotransmitter metabolism (Prandovszky et al., 2011), metabolic imbalances (Myint et al., 2011), anti-inflammatory activation (Meyer, 2011). Even seemingly unrelated factors such as modulation from exercise (Gleeson et al., 2011), neurocognitive training (Eack et al., 2010), or various types of stress (see for general interactions Dantzer et al., 2008) may relate to LLNI. 8.5. Links between CNS and systemic factors Surprisingly, direct interactions exist between systemic immunity and the CNS (Saxton et al., 2011). For example, commensal microbiota may trigger autoimmune demyelination (Willyard, 2011) or in other ways interact with CNS autoimmunity (Dunne and Cooke, 2005, Grace et al., 2011). Treatments may also interact through various mechanisms with the immune response itself (Martins-de-Souza et al., 2011, Nikisch et al., 2011, Piontkewitz et al., 2009). That such interactions have in vivo relevance was demonstrated by the use of psychopharmaca in experimental autoimmune encephalitis (Musgrave et al., 2011). Pathomechanisms involved in infections include size, geometry, kinetics of molecular patterns (Bachmann and Jennings, 2010), and others (see, also, BDV infection discussed herein). 8.6. Multiple sclerosis The ME scenario may, in various aspects, be similar to the rather well-investigated case of MS where a role of risk genes is proven (Sawcer et al., 2011) as several infectious agents seem to represent triggers (compared with Bostrom et al., 2009, McLeod et al., 2011, Wallin et al., 2009). Thus, the disease is considered an autoimmune disease. Pathogenetic details were extensively studied in experimental models, mainly experimental autoimmune encephalitis (EAE), and the best model represents Theiler's virus infection (Oleszak et al., 2004). Over time, complex pathogenetic events emerge, as represented in 83 EAE and in MS brains. Immune responses involving complex inflammatory mechanisms and various arms of the immune system emerge even locally within the CNS (Krishnamoorthy et al., 2009, Meinl et al., 2006, Skulina et al., 2004). It is noted that psychiatric symptoms are frequent in MS, including rare schizophrenia type psychoses (Gross and Huber, 2007), and brain imaging findings demonstrated considerable similarities in between MS and schizophrenia subgroup (Davis et al., 2003). 8.7. How do specific schizophrenic symptoms emerge? Neuroinflammation may lead to a range of psychiatric syndromes along pathways from exposure to chronic illness or disability, which are difficult to disentangle because they are always subject to change depending on various human behavioral traits (O'Connor et al., 2006). The involvement of genes in a broad picture of abnormalities in schizophrenia has been recognized (Verrall et al., 2010). Why, in the case of LLNI situations or in classical CNS inflammation in one individual, schizophrenia-like psychosis develop, whereas other psychiatric syndromes develop in other cases, remains nevertheless unanswered. Beyond pre-existing liability factors, especially genetic ones, in the neurodevelopmental presentation as an endophenotype and for various downstream factors emerging over time, neurotransmitter functioning may likely be involved and plausibly rather specific symptoms may easily emerge at these levels of the CNS functional system, and in addition depend from the topology of pathology (this aspect has been in part discussed in the neuroimaging section, more to be found in a neurophysiological and functional imaging field not reviewed here). Risk genes involved in the schizophrenia subgroup were expressed in human induced pluripotent stem cells from schizophrenic patients (Brennand et al., 2011). As these single risk genes were mainly involved in neuronal functioning, they may preferably contribute to the type of symptoms evolving but not necessarily to the disease process itself. From this perspective, these genes should similarly be involved in MS associated or other ‘organic’ schizophrenic type psychoses, which remains to be shown. In sum, the ME scenario provides a multitude of immune pathological mechanisms and states involved in LLNI and influenced by the interaction between genes and environment, thereby shaping an emerging immune inflammatory response over time. Neurotropic infections have the potential to induce numerous pathologies and downstream events ranging from neurodevelopmental aberrations to LLNI and classical neuroinflammation and even including infection-induced cellular dysfunction on the molecular level. Infections may trigger LLNI, which may occur at many time points during the lifetime of an individual. At certain ages, an individual is either more or less sensitive to harmful specific infections, several of which present with high pathogenicity at ages of preferred disease onset; this is a finding that seems of special interest for research on the meS subgroup. Reactivation, autoimmunity or infection-induced molecular pathomechanisms may be involved. CNS dysfunctions relating to these various mechanisms may emerge and change over time, influenced by the type of the endogenous CNS immune response in interaction with systemic immunity. Major sites of CNS endogenous immune responses represent the ECF and the CSF spaces and, thus, involve the whole brain at many sites. However, the primary triggers of LLNI, such as infections, autoimmunity, toxicity or trauma, are difficult to demonstrate because they may not be present after the onset of the disease, and in the case of persistent infections, they are generally difficult to detect. A teaching example for the research situation in schizophrenia may represent general autoimmune disorders in which three major factors are required: genes, environment and immune system. The course characteristics of autoimmune disorders resemble, in various ways, the course characteristics of schizophrenia, such as prodrome, variability, transition, non-transition, relapse, chronicity, and defect. When accepting the terminology of a meS subgroup, one might further differentiate primary from secondary meS. Nevertheless, LLNI 84 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 may also be involved in neurodevelopmental schizophrenia, e.g., as a late hit. Both unspecificity and convergence of pathomechanisms are not unusual as different etiologies produce surprisingly similar diseases, e.g., similar syndromal outcome from heterogeneous genetic causes (Auerbach et al., 2011) or molecular pathology from genetic and non-genetic aspects converging to finally relevant pathomechanisms (Voineagu et al., 2011). Persisting infections may play a more important role than presently assumed. Diagnostic difficulties with persistent infections are not singular to the neuropsychiatric field, as evidenced from a resume about the present standing of research into tuberculosis. Although this research was well funded over many years because of its enormous impact on human health, the methods remained insensitive and clinical diagnoses remained difficult regarding the individual case requiring a broad array of diagnostic approaches, including a century old sputum microscopy (Lawn and Zumla, 2011). These issues were compounded by the diagnostic challenge of tuberculous meningitis. Similar difficulties of diagnosis can be found in late lyme neuroborreliosis (compared with Fallon et al., 2009, Stanek et al., 2012) and in prodromal stages to HIVe or LE. In such contexts, the findings suggesting the prevalence of a meS subgroup appear promising. 9. The peripheral CSF outflow pathway and the anatomy of schizophrenia The explanations of the pathophysiology of schizophrenia, not the rarely preferred simplified views of a chronic stable disease, focused on synaptic pathologies to explain the entire disease. In fact, symptoms and underlying dysfunctions in schizophrenia are apparently widely distributed and involve both the gray and white matter, in other words, the whole brain. In addition, it appeared that both peripheral and central neuronal dysfunction underlie the symptoms observed when investigating for olfactory dysfunction (review see Bechter, 2011). Dysautonomia is also frequent in psychoses, including schizophrenia, which seems related to central dysfunction but may include a peripheral site of neuronal dysfunction (Bar et al., 2004, 2010). The same seems true for abnormal pursuit eye tracking (compared with Lencer et al., 2011). Such parallelism of peripheral and central neuronal dysfunction must be explained and could, for example, relate to cellular receptors or cellular metabolic dysfunction expressed at the central and peripheral sites. Furthermore, systemic pathologies, such as proteome abnormalities (Bahn and Schwarz, 2011) or activated blood monocytes (Drexhage et al., 2010b), could mediate such peripheral and central dysfunction. But there were also some unexplained findings such as subtle muscle lesions that resembled lesions found in meningoencephalitis apparent in approximately 50% of the cases (Meltzer and Crayton, 1974). The muscle lesions could have been replicated and, thus, were speculatively attributed to a genetic abnormality (Flyckt et al., 2000). In addition to systemic abnormalities and central and peripheral neuronal dysfunction, the underlying pathogenetic principle must be explained by the change in brain structure and function over time in all aspects. A genetic aberration could, in general, underlie such a situation as observed in the monoamine neurotransmitter disorders, preferably when genes were related to basic cellular functions. However, in nS, one should identify the time trajectories of abnormal neurodevelopment, which was incomplete (see above). When nS represented a major subgroup, such as the prevailing hypothesis of schizophrenia, one would expect that as in monoamine neurotransmitter disorders, several types of nS would be found and symptom patterns would vary such that disease onset would relate to a certain developmental period in which the respective etiology was active. In detailed studies of symptom patterns in different schizophrenia risk groups, each of the subgroups presented a similar distribution of symptom patterns (Ribbe et al., 2011); this finding is not consistent with the idea of neurodevelopmental schizophrenia. According to the watershed model, the risk genes involved in schizophrenia are proposed to be few (Brennand et al., 2011); this, however, has yet to be proven and may not explain causality of disease (see above). Distributed CNS pathology and dysfunction involving gray and white matter is, together, often termed disconnectivity and confirmed from various approaches including neuroimaging (see above), histopathology (Schmitt et al., 2011a), and neurophysiology (Foxe et al., 2011, Mulert et al., 2011a, 2011b, Strik et al., 2002) or from combined approaches (Heuser et al., 2011). Explanations of this complexity of aberrations arising in parallel at various sites and systems of the CNS remain preliminary (see, for example, Andreone et al., 2007 and the discussions herein). Influences from psychopharmacological treatment, although demonstrated, represent a negligible aspect with regard to understanding the disease (de Castro-Manglano et al., 2011). The experimental findings for the VT mode were corroborated in the meanwhile (Agnati et al., 2010, Fuxe et al., 2010), representing an important physiological pathway with a consistent importance of the flow of the ECF throughout the brain moved by the pulsating brain with each cardiac cycle, and involving, in parallel, the CSF flow (compared with Bechter, 2011, Fuxe et al., 2010, Gupta et al., 2009, 2010). The PCOP pathway (Bechter, 2011) was first determined to be important in immunology in that CNS antigens are released along this pathway into the periphery to induce CNS-specific immune responses (Cserr and Knopf, 1992). In research approach cases, therapy-resistant depression or schizophrenia was improved with cerebrospinal fluid filtration (Bechter et al., 1999, 2000) before being found to be an effective treatment of neurological autoimmune disorder Guillain–Barré syndrome (Wollinsky et al., 2001). Clinical observations during CSF filtration suggested a direct role of CSF signaling within the subarachnoid spaces and at peripheral sites and initiated the PCOP hypothesis, which could be relevant for psychoses, including schizophrenia, and for neurodegenerative and neuroinflammatory disorders in general (Bechter, 2011), although this remains highly speculative. When assuming that sites of the pathway itself may be involved in pathology from CSF signaling along brain nerves and peripheral nerves and even into the tissues connected by the nerves (Bechter, 2011), many aspects of the PCOP hypothesis must be further studied. Yet known is for example, that CSF cells are trafficking into the periphery along the PCOP, as demonstrated in experiments at the cribriform plate (Goldmann et al., 2006, Locatelli et al., 2012) and in humans along the peripheral nerves in a first case (Schmitt et al., 2011b). Interestingly, the role of CSF cells was previously underestimated with respect to their apparent importance for CNS immunity in general (Schwartz and Kipnis, 2011, Schwartz and Shechter, 2010) and their rather direct influence on cognitive functions (Derecki et al., 2010). 10. Conclusion This update was given with a personal background of clinical experience of more than 30 years in psychiatry that included continuous responsibility of a specialized ward for young schizophrenic patients with complicated and therapy-resistant courses. Individual cases of schizophrenia have been personally followed for more than 30 years and have been found to demonstrate surprising unpredictability and variance of courses. At the extremes, one case of initially therapy-resistant psychosis was successfully treated and after one year never relapsed during about 30 years. At the other extreme, cases of devastating courses within a few years, or the rare case that began with schizophrenia and after 10 years presented as multiple sclerosis with unusual course aspects. Gerd Huber and co-workers showed these extremes in a milestone study over the long term (Huber et al., 1979) demonstrating an extraordinary variation of courses then differentiated in twelve subtypes. Such course variability was broadly confirmed in a number of studies in the meanwhile though subtyping was not introduced in the established diagnostic systems. The recent views from the scientific K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 field on schizophrenia indicate there is only a partial understanding of the disorder (see Table 5). Accordingly, there is a strong argument against a revival of a simplistic all inclusive hypothesis. An approach of etiopathogenetic subgrouping of schizophrenia according to the major constraints of results (as given in Table 4) appears preferable as the overwhelming consensus in research is that schizophrenia represents a heterogeneous disorder. The developments in medicine in general and in psychiatry specifically, e.g., the autism spectrum disorders, which have often been mentioned with an overlap and similarity to schizophrenic disorders, clearly argue against the view that one size fits all or that there may be a single appropriate approach. Furthermore, the unspecificity of psychiatric symptoms is a well-established phenomenon, implicitly suggesting that the heterogeneity of etiologies is just the other side of one coin. Nevertheless, to develop criteria for an appropriate subgrouping of schizophrenia of different etiopathogenetic types of schizophrenia will remain a challenge for psychiatric research, especially when developing consensus criteria. Infectious agents are risk factors for schizophrenia, usually explained in a developmental scenario (Brown, 2011a). However, a late hit can hardly been explained by aberrant neurodevelopment, if not directly involving immune mechanisms, and a recent milestone study from Denmark was surprisingly well compatible with the ME scenario: both, severe infections and autoimmune disorders during lifetime, increase additively and non-specifically the risk of psychoses including schizophrenia (Benros et al., 2011). For the meS subgroup proposed here, this update of the ME hypothesis demonstrates an emerging knowledge about LLNI situations that prevail within the CNS, widening the basis for the assumption that LLNI may underlie a variety of psychiatric disorders, especially subgroups of affective and schizophrenic type psychoses. The evidence of overlap between affective and schizophrenic disorders with regard to risk genes and, therefore, disease pathogenesis is strongly corroborated from large genetic studies and was well compatible with the longknown unspecificity of organic etiologies, including encephalitis for psychoses. LLNI situations differ from classical neuroinflammation in that LLNI represents a low degree inflammation but may nevertheless be able to induce psychiatric disorders including schizophrenia emerging within a complex pathogenetic scenario. Such a perspective was corroborated from findings in experimental neuroimmunology and from research in newly detected CNS inflammatory disorders, especially limbic encephalitis where during the prodromal phase, various psychiatric syndromes prevail though classical CNS inflammation has yet to be found. Many researchers consider schizophrenia to represent a heterogeneous disorder, and broad consensus was found, especially within the neuroimaging literature, that a so-called late hit is required to explain the observed pathologies. Thus, something must happen to or occur in the CNS around the critical disease phases from prodrome to disease onset and especially during the first years after onset. This hit apparently leads to considerable disturbances of brain function and even brain structure. However, the quality of the hit remained undefined. Many aspects of the findings in neuroimaging and in neuropathology match the view that this hit could be represented by an LLNI process. LLNI pathomechanisms could explain core aspects of the observed pathology through the type of the pathomechanism involved and the specific topological issues, i.e., the widely distributed brain dysfunction. The brain barriers (blood–brain barrier, blood–CSF barrier, meningeal–CSF barrier) and respective interactions among the bodily compartments involved and the CSF cells play a more important role in CNS functioning than previously assumed. Such new insights align well with the ME hypothesis. Disturbances mediated by extra-cellular CNS fluid and CSF, together in exchange with the systemic blood compartment, have the potential to explain, by a common pathogenetic link like LLNI, major findings in schizophrenia by anatomy, especially the ‘non-localization’ (see above), meaning anatomically widely distributed pathologies, and possible various pathophysiologies, which include distributed brain 85 dysfunction; emerging disturbed brain structure including slight atrophy involving both gray and white matter, course variability resembling the course variability of autoimmune disorders (including the prodromal phase, transition, non-transition, chronicity, relapse, processactive stages and defect). The parallelism between central and peripheral neural dysfunction in schizophrenia, e.g., the olfactory system, or the findings of aberrant proteomics and of monocytes activation in peripheral blood, clearly suggests that schizophrenia should, in part, be understood as a systemic disease with a preferential involvement of the brain rather than being considered an exclusive brain disease. The pathways and mechanisms of the bidirectional pathways between the brain and the peripheral immune system are important and previously unknown mechanisms become evident (Anthony et al., 2012). The majority of schizophrenia cases seem related to complex genetic traits, which is a scenario resembling that found in autoimmune disorders, because a number of infectious agents seem to be involved. Important epidemiological aspects of schizophrenia, such as the preferred age of onset, the continuous high risk gene pool within populations despite low numbers of progeny and the role of infections (age-related variance of pathogenicity), would well align with the ME hypothesis. General autoimmune disorders require three factors (gene, environment, immune system), thus constituting a paradigm for the meS subgroup. Many infections present with overall low pathogenicity within the populations, which is consistent with the ME hypothesis, though it is difficult to research because the contribution of a single agent is hidden and dependent of interacting factors. The infectious agent may, nevertheless, represent the conditio sine qua non (needed factor), e.g., see the recent story with gastric ulcer and helicobacter pylori infection. An overlap between schizophrenia and other psychiatric syndromes produced from similar LLNI causes is expected as the meS subgroup presumably links to certain genetic and other pre-existing liability factors. A recent CSF study performed according to the advanced CSF analytic methods, validated in thousands of patients mainly from neurology, demonstrated LLNI being most likely present in a subgroup of approximately 40% of the therapy-resistant cases of schizophrenia. With more sensitive methods to assess LLNI, the size of this subgroup will likely be larger based on a preliminary study with the newly detected autoantibodies discussed in LE research (Steiner et al., 2011). The schizophrenia research field is a leading field in psychiatric research since the decennia with regard to developing refined methods and with regard to epidemiological approaches. However, the research field has not, to date, been successful in subgrouping the etiopathogenesis. Although less research effort has been put into the research of organic psychiatric disorder when presenting as nondementive syndromes, these rarer organic psychotic and nonpsychotic, i.e., personality, disorders could promote the development of valid diagnostic criteria for schizophrenia and other psychiatric disorders with an organic etiopathogenesis or partial organic contribution. However, the accepted diagnostic systems, DSM IV and ICD-10, do not provide operational criteria to classify organic psychiatric disorders, but rather rely on a formula that is applied to a clinician's evaluation criteria. This is a preliminary level of categorization, although in organic psychiatric disorders, this is the best available possibility for evaluating and strictly defining criteria. For some single cases of personality disorder or therapy-resistant depression, the appropriate ‘organic’ classification has been exemplified: a case of possible streptococcal infection associated autoimmune-related therapy-resistant depression was classified as probable ‘organic’ when including the therapeutic effect from penicillin treatment plus a follow-up of five years that included the results from neuroimaging and laboratory testing and CSF investigations (Bechter et al., 2007). Cases of possible arachnoid cysts associated personality disorders were classified as probable when rapid improvement after neurosurgical treatment of arachnoid cysts was noted. It was, as a consequence, suggested to 86 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 revise the current weak operational criteria (Bechter et al., 2010b). Apparently, there is a need to improve consensus criteria for nondementive organic psychiatric disorders as a general challenge for psychiatry including schizophrenic and other psychiatric disorders with partial ‘organic’ aspects in pathogenesis (Bechter, in press). The major challenge, therefore, is the improved categorization of the findings in an improved diagnostic approach, which should include the major compartments involved in disease pathogenesis, i.e., the brain itself, blood as a systemic indicator, and, presently underscored, the CSF. The importance of combining these three major bodily compartments in the diagnostic approach is illustrated in the autism spectrum disorders where it is expressively claimed that even in genetically caused autism spectrum disorders, CSF investigation is needed in addition to other approaches for a correct differential diagnosis (see above). More detailed proposals for the diagnostic categorization in this respect cannot be made in this review because it would require a careful discussion of the validity of methods, limits of sensitivity, and the immanently more general limitation of the approaches to understand brain diseases, apparently often only valid when combining many approaches and findings. With improved diagnostic methods, improved therapeutic success is expected. In fact, several indications are available that suggest the therapeutic effects related to neuroprotective and anti-inflammatory aspects along with anti-inflammatory medications are helpful (not reviewed here). As etiologies, pathophysiological details and, over time, the type of LLNI pathomechanisms may vary, the preferred research and diagnostic approach is at the individual case level. New specific methods that are more sensitive to detecting LLNI are required but should be based and validated by the longstanding clinical experience available in the neurological and, especially, CSF research field. The number of mechanisms most likely involved in an LLNI situation is constantly increasing, which makes the situation for research attractive and challenging. With regard to the neurodevelopmental hypothesis of schizophrenia, it was recommended to carefully observe the expertise in neighboring fields, e.g., in developmental cognitive neuroscience (Bunge, 2012) or neurodevelopmental epilepsy (Bozzi et al., 2012), to differentiate the possible neurodevelopmental insults onto the brain with regard to their variant syndromal outcome. With regard to the meS subgroup, new insights into the role of endogenous viruses in evolution and host biology seems interesting which demonstrate a close interaction (and continuous battle) between viruses and hosts, including many conventional viruses (Feschotte and Gilbert, 2012). New views about the role that immunity plays in multiple sclerosis (Locatelli et al., 2012) are surely insightful as MS represents the best studied inflammatory CNS disorder in humans. The recent insight that inflammation in experimental autoimmune encephalitis is beginning at the meninges (see above) was confirmed in human multiple sclerosis when investigating biopsy material: the results contradicted the previous assumption that cortical atrophy was non-inflammatory, instead appeared associated with short-lived inflammation (Lucchinetti et al., 2011). A similar scenario of short-lived classical inflammation hardly to be detected, was assumed in the ME hypothesis and exemplified for BDV infection models, nevertheless recognizing that other types of LLNI may prevail in meS subgroup (compared with Bechter, 2001). In conclusion, psychiatry's need to adopt a highly active and growing psychoimmunology approach to psychiatric disorders which has led to some fascinating results (Licinio, 2010) (compare the abstracts of the 11th Psychoimmunology Expert Meeting March 2012), is promoted here. For example, one small, pioneering but growing field with interdisciplinary views is the specific approach of HERV-W elements, which examines its relevance in MS and in schizophrenia (Leboyer et al., 2011, Li et al., 2011). In a second example, a recent milestone study from Denmark shows the additive effect of infections and autoimmune disorders during life time on psychosis risk (Benros et al., 2011). In a third example, this is the emerging findings from CSF studies indicating LLNI to prevail in a schizophrenia subgroup (Bechter, 2011, Maxeiner et al., 2009, Oleszak et al., 2007), and in a fourth example the imaging of the underlying neuroinflammatory process by microglia activation or indirect signs (in the interpretation from an ME perspective!) of structural abnormalities (see above), or progressive membrane phospholipid change in first episode schizophrenia with gradual inclusion of brain regions with an initial increase followed by a decrease of metabolites (Miller et al., 2012). As the details of these studies demonstrate that change is often observed the method one size fits all may not be successful in such a complex disease as schizophrenia represents instead the diagnostic approach should be based on strict rules in a broad clinical validate experience of possibilities and limitations of these methods and combining various approaches in one case in both, research and, when validated, in individual cases as is the typical approach to the clinical medicine and why not in subgrouping schizophrenia. The recent controversy in limbic encephalitis research seems to demonstrate that the discussion outlined here with the updated ME hypothesis is at the heart of the problem: in clinically strictly defined limbic encephalitis the neuroimaging and CSF findings are very sensitive and the clinical patterns found match with the type of autoantibodies (Dalmau, 2012), whereas in schizophrenia subgroup of antibody-mediated encephalitis both, neuroimaging and CSF investigation, may not be sensitive and serum investigation was considered to be possibly sufficient (Lennox et al., 2012); however, there is an ongoing debate, e.g. whether aggressive treatments as used in strictly defined limbic encephalitis may be justified or not in schizophrenia subgroup presenting with CNS autoantibodies just in serum. Acknowledgment For continuous support of the CSF studies and studies on the elucidation of possible role of BDV for psychiatric disorders we thank the Margarete-Ammon-Stiftung (Munich). References Adamson LA, Fowler LJ, Clare-Salzler MJ, Hobbs JA. Parvovirus B19 infection in Hashimoto's thyroiditis, papillary thyroid carcinoma, and anaplastic thyroid carcinoma. Thyroid 2011;21:411–7. Agnati LF, Bjelke B, Fuxe K. Volume versus wiring transmission in the brain: a new theoretical frame for neuropsychopharmacology. Med Res Rev 1995;15:33–45. Agnati LF, Guidolin D, Guescini M, Genedani S, Fuxe K. Understanding wiring and volume transmission. Brain Res Rev 2010;64:137–59. Allen JE, Maizels RM. Diversity and dialogue in immunity to helminths. Nat Rev Immunol 2011;11:375–88. Andreasen N. Neuroimaging of schizophrenia: commentary. In: Shenton ME, Turetsky BI, editors. Understanding neuropsychiatric disorders — insights from neuroimaging. New York: Cambridge University Press; 2011. p. 88. Andreasen NC, Nopoulos P, Magnotta V, Pierson R, Ziebell S, Ho BC. Progressive brain change in schizophrenia: a prospective longitudinal study of first-episode schizophrenia. Biol Psychiatry 2011;70(7):672–9. Andreone N, Tansella M, Cerini R, Versace A, Rambaldelli G, Perlini C, et al. Cortical white-matter microstructure in schizophrenia. Diffusion imaging study. Br J Psychiatry 2007;191:113–9. Anthony DC, Couch Y, Losey P, Evans MC. The systemic response to brain injury and disease. Brain Behav Immun 2012;26:534–40. Arbuckle JH, Medveczky MM, Luka J, Hadley SH, Luegmayr A, Ablashi D, et al. The latent human herpesvirus-6A genome specifically integrates in telomeres of human chromosomes in vivo and in vitro. Proc Natl Acad Sci U S A 2010;107:5563–8. Arias I, Sorlozano A, Villegas E, Luna JD, McKenney K, Cervilla J, et al. Infectious agents associated with schizophrenia: a meta-analysis. Schizophr Res 2012;136(1–3): 128–36. Arnold IC, Dehzad N, Reuter S, Martin H, Becher B, Taube C, et al. Helicobacter pylori infection prevents allergic asthma in mouse models through the induction of regulatory T cells. J Clin Invest 2011;121:3088–93. Auerbach BD, Osterweil EK, Bear MF. Mutations causing syndromic autism define an axis of synaptic pathophysiology. Nature 2011;480:63–8. Bachmann MF, Jennings GT. Vaccine delivery: a matter of size, geometry, kinetics and molecular patterns. Nat Rev Immunol 2010;10:787–96. Bahn S, Schwarz E. Serum-based biomarkers for psychiatric disorders. Nervenarzt 2011;82:1395–403. Bar KJ, Greiner W, Jochum T, Friedrich M, Wagner G, Sauer H. The influence of major depression and its treatment on heart rate variability and pupillary light reflex parameters. J Affect Disord 2004;82:245–52. K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 Bar KJ, Berger S, Metzner M, Boettger MK, Schulz S, Ramachandraiah CT, et al. Autonomic dysfunction in unaffected first-degree relatives of patients suffering from schizophrenia. Schizophr Bull 2010;36:1050–8. Bartholomaus I, Kawakami N, et al. Effector T cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions. Nature 2009;462(7269):94–8. Bayer TA, Falkai P, Maier W. Genetic and non-genetic vulnerability factors in schizophrenia: the basis of the “two hit hypothesis”. J Psychiatr Res 1999;33:543–8. Bechmann I, Galea I, Perry VH. What is the blood–brain barrier (not)? Trends Immunol 2007;28:5-11. Bechter K, Bogerts B. Inflammatory and neuroimmunological aspects in psychiatric disorders. Highlights of the 9th Psychoimmunology Expert Meeting: Neuropsychoimmunology of Psychoses. Immune and Inflammatory Aspects of Psychoses. In Vivo 2007;21:917–23. Bechter K. Borna Disease Virus. Mögliche Ursache neurologischer und psychiatrischer Störungen des Menschen. In: Hippius H, Janzarik W, Müller C, editors. Monographien aus dem Gesamtgebiete Psychiatrie. Darmstadt: Steinkopff; 1998. Bechter K. Mild encephalitis underlying psychiatric disorder — a reconsideration and hypothesis exemplified on Borna disease. Neurol Psychiatry Brain Res 2001;9:55–70. Bechter K. The peripheral cerebrospinal fluid outflow pathway — physiology and pathophysiology of CSF recirculation: a review and hypothesis. Neurol Psychiatry Brain Res 2011;17:51–66. Bechter, K., in press. Diagnosis of infectious or inflammatory psychosyndromes. Open J Neurol. Bechter K, Hodgkiss A. Research strategies in ‘slow’ infections in psychiatry. Hist Psychiatry 1995;6:503–11. Bechter K, Herzog S, Behr W, Schuttler R. Investigations of cerebrospinal fluid in Borna disease virus seropositive psychiatric patients. Eur Psychiatry 1995;10:250–8. Bechter K, Herzog S, Schreiner V, Wollinsky KH, Schüttler R. Cerebrospinal fluid filtration in a case of schizophrenia related to ‘subclinical’ Borna disease virus encephalitis. In: Müller N, editor. Psychiatry, psychoneuroimmunology and viruses. Key top brain res. Wien: Springer; 1999. p. 19–35. Bechter K, Herzog S, Schreiner V, Brinkmeier H, Aulkemeyer P, Weber F, et al. Borna disease virus-related therapy-resistant depression improved after cerebrospinal fluid filtration. J Psychiatr Res 2000;34:393–6. Bechter K, Bindl A, Horn M, Schreiner V. Therapieresistente Depression mit Fatigue. Fall einer vermutlichen streptokokkenassoziierten Autoimmunkrankheit. Nervenarzt 2007;78(338):340–1. Bechter K, Reiber H, Herzog S, Fuchs D, Tumani H, Maxeiner HG. Cerebrospinal fluid analysis in affective and schizophrenic spectrum disorders: identification of subgroups with immune responses and blood–CSF barrier dysfunction. J Psychiatr Res 2010a;44:321–30. Bechter K, Wittek R, Seitz K, Antoniadis G. Personality disorders improved after arachnoid cyst neurosurgery, then rediagnosed as ‘minor’ organic personality disorders. Psychiatry Res 2010b;184:196–200. Belarbi K, Arellano C, Ferguson R, Jopson T, Rosi S. Chronic neuroinflammation impacts the recruitment of adult-born neurons into behaviorally relevant hippocampal networks. Brain Behav Immun 2011;26:18–23. Belyi VA, Levine AJ, Skalka AM. Unexpected inheritance: multiple integrations of ancient bornavirus and ebolavirus/marburgvirus sequences in vertebrate genomes. PLoS Pathog 2010;6:e1001030. Benedetti F, Radaelli D, Poletti S, Falini A, Cavallaro R, Dallaspezia S, et al. Emotional reactivity in chronic schizophrenia: structural and functional brain correlates and the influence of adverse childhood experiences. Psychol Med 2011;41:509–19. Benros M, Nielsen P, Nordentoft M, Eaton WW, Dalton SO, Mortensen PB. Autoimmune diseases and severe infections as risk factors for schizophrenia — a 30-year population-based register study. Am J Psychiatry 2011;168:1303–10. Bentivoglio M, Mariotti R, Bertini G. Neuroinflammation and brain infections: historical context and current perspectives. Brain Res Rev 2011;66:152–73. Bertsias GK, Boumpas DT. Pathogenesis, diagnosis and management of neuropsychiatric SLE manifestations. Nat Rev Rheumatol 2010;6:358–67. Bien CG, Urbach H, Schramm J, Soeder BM, Becker AJ, Voltz R, et al. Limbic encephalitis as a precipitating event in adult-onset temporal lobe epilepsy. Neurology 2007;69: 1236–44. Billich C, Sauder C, Frank R, Herzog S, Bechter K, Takahashi K, et al. High-avidity human serum antibodies recognizing linear epitopes of Borna disease virus proteins. Biol Psychiatry 2002;51:979–87. Bode L, Ludwig H. Borna disease virus infection, a human mental-health risk. Clin Microbiol Rev 2003;16:534–45. Bondy B. Genetics in psychiatry: are the promises met? World J Biol Psychiatry 2011;12:81–8. Bostrom I, Callander M, Kurtzke JF, Landtblom AM. High prevalence of multiple sclerosis in the Swedish county of Varmland. Mult Scler 2009;15:1253–62. Bozzi Y, Casarosa S, Caleo M. Epilepsy as a neurodevelopmental disorder. Front Psychiatry 2012;3. Article 19. Braak H, Del Tredici K. Invited article: nervous system pathology in sporadic Parkinson disease. Neurology 2008;70:1916–25. Brennand KJ, Simone A, Jou J, Gelboin-Burkhart C, Tran N, Sangar S, et al. Modelling schizophrenia using human induced pluripotent stem cells. Nature 2011;473:221–5. Bromet EJ, Kotov R, Fochtmann LJ, Carlson GA, Tanenberg-Karant M, Ruggero C, et al. Diagnostic shifts during the decade following first admission for psychosis. Am J Psychiatry 2011;168:1186–94. Brown AS. Further evidence of infectious insults in the pathogenesis and pathophysiology of schizophrenia. Am J Psychiatry 2011a;168:764–6. Brown AS. The environment and susceptibility to schizophrenia. Prog Neurobiol 2011b;93:23–58. Brown AS. Exposure to prenatal infection and risk of schizophrenia. Front Psychiatry 2011c;2:63. 87 Brown AS, McGrath JJ. The prevention of schizophrenia. Schizophr Bull 2011;37:257–61. Brown AS, Cohen P, Harkavy-Friedman J, Babulas V, Malaspina D, Gorman JM, et al. A.E. Bennett Research Award. Prenatal rubella, premorbid abnormalities, and adult schizophrenia. Biol Psychiatry 2001;49:473–86. Brown AS, Begg MD, Gravenstein S, Schaefer CA, Wyatt RJ, Bresnahan M, et al. Serologic evidence of prenatal influenza in the etiology of schizophrenia. Arch Gen Psychiatry 2004;61:774–80. Bucy DS, Brown MS, Bielefeldt-Ohmann H, Thompson J, Bachand AM, Morges M, et al. Early detection of neuropathophysiology using diffusion-weighted magnetic resonance imaging in asymptomatic cats with feline immunodeficiency viral infection. J Neurovirol 2011;17:341–52. Buka SL, Tsuang MT, Torrey EF, Klebanoff MA, Bernstein D, Yolken RH. Maternal infections and subsequent psychosis among offspring. Arch Gen Psychiatry 2001;58:1032–7. Bunge SA. The developing human brain: a frontiers research topic. Front Hum Neurosci 2012;6:1. Bustillo JR, Rowland LM, Mullins P, Jung R, Chen H, Qualls C, et al. 1H-MRS at 4 Tesla in minimally treated early schizophrenia. Mol Psychiatry 2010;15:629–36. Cadwell K, Patel KK, Maloney NS, Liu TC, Ng AC, Storer CE, et al. Virus-plus-susceptibility gene interaction determines Crohn's disease gene Atg16L1 phenotypes in intestine. Cell 2010;141:1135–45. Calabrese M, Agosta F, Rinaldi F, Mattisi I, Grossi P, Favaretto A, et al. Cortical lesions and atrophy associated with cognitive impairment in relapsing–remitting multiple sclerosis. Arch Neurol 2009;66:1144–50. Carbone KM. Borna disease virus and its role in neurobehavioral disease. Washington, DC: ASM Press; 2002. Carlsson ML, Carlsson A, Nilsson M. Schizophrenia: from dopamine to glutamate and back. Curr Med Chem 2004;11:267–77. Caspi A, Hariri AR, Holmes A, Uher R, Moffitt TE. Genetic sensitivity to the environment: the case of the serotonin transporter gene and its implications for studying complex diseases and traits. Am J Psychiatry 2010;167:509–27. Chawarska K, Campbell D, Chen L, Shic F, Klin A, Chang J. Early generalized overgrowth in boys with autism. Arch Gen Psychiatry 2011;68:1021–31. Chen GY, Nunez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol 2010;10:826–37. Cherner M, Cysique L, Heaton RK, Marcotte TD, Ellis RJ, Masliah E, et al. Neuropathologic confirmation of definitional criteria for human immunodeficiency virus-associated neurocognitive disorders. J Neurovirol 2007;13:23–8. Chevalier G, Suberbielle E, Monnet C, Duplan V, Martin-Blondel G, Farrugia F, et al. Neurons are MHC class I-dependent targets for CD8 T cells upon neurotropic viral infection. PLoS Pathog 2011;7:e1002393. Clarke MC, Tanskanen A, Huttunen M, Leon DA, Murray RM, Jones PB, et al. Increased risk of schizophrenia from additive interaction between infant motor developmental delay and obstetric complications: evidence from a population-based longitudinal study. Am J Psychiatry 2011;168:1295–302. Clemente R, Sisman E, Aza-Blanc P, de la Torre JC. Identification of host factors involved in Borna disease virus cell entry through a small interfering RNA functional genetic screen. J Virol 2010;84:3562–75. Coban O, Bahar S, Akman-Demir G, Tasci B, Yurdakul S, Yazici H, et al. Masked assessment of MRI findings: is it possible to differentiate neuro-Behcet's disease from other central nervous system diseases? [corrected]. Neuroradiology 1999;41: 255–60. Cohen IR. Real and artificial immune systems: computing the state of the body. Nat Rev Immunol 2007;7:569–74. Cserr HF, Knopf PM. Cervical lymphatics, the blood–brain barrier and the immunoreactivity of the brain: a new view. Immunol Today 1992;13:507–12. Daito T, Fujino K, Watanabe Y, Ikuta K, Tomonaga K. Analysis of intracellular distribution of borna disease virus glycoprotein fused with fluorescent markers in living cells. J Vet Med Sci 2011a;73:1243–7. Daito T, Fujino K, Honda T, Matsumoto Y, Watanabe Y, Tomonaga K. A novel Borna disease virus vector system that stably expresses foreign proteins from an intercistronic noncoding region. J Virol 2011b. Dalman C, Allebeck P, Gunnell D, Harrison G, Kristensson K, Lewis G, et al. Infections in the CNS during childhood and the risk of subsequent psychotic illness: a cohort study of more than one million Swedish subjects. Am J Psychiatry 2008;165: 59–65. Dalmau J. Anti-NMDAR and other synaptic mechanisms of autoimmune encephalitis. Abstracts of the 8th International Congress on Autoimmunity; 2012. www.kenes.com/ autoimmunity. Dalmau J, Lancaster E, Martinez-Hernandez E, Rosenfeld MR, Balice-Gordon R. Clinical experience and laboratory investigations in patients with anti-NMDAR encephalitis. Lancet Neurol 2011;10:63–74. Dantzer R, Capuron L, Irwin MR, Miller AH, Ollat H, Perry VH, et al. Identification and treatment of symptoms associated with inflammation in medically ill patients. Psychoneuroendocrinology 2008;33:18–29. Davis KL, Stewart DG, Friedman JI, Buchsbaum M, Harvey PD, Hof PR, et al. White matter changes in schizophrenia: evidence for myelin-related dysfunction. Arch Gen Psychiatry 2003;60:443–56. de Castro-Manglano P, Mechelli A, Soutullo C, Gimenez-Amaya J, Ortuno F, McGuire P. Longitudinal changes in brain structure following the first episode of psychosis. Psychiatry Res 2011;191:166–73. De Hert M, Wampers M, Jendricko T, Franic T, Vidovic D, De Vriendt N, et al. Effects of cannabis use on age at onset in schizophrenia and bipolar disorder. Schizophr Res 2011;126:270–6. De Nayer AR, Myant N, Sindic CJ. A subacute behavioral disorder in a female adolescent. Autoimmune anti-N-methyl-D-aspartate receptor encephalitis associated with ovarian teratoma. Biol Psychiatry 2009;66:e13–4. 88 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 Dean K, Fearon P, Morgan K, Hutchinson G, Orr K, Chitnis X, et al. Grey matter correlates of minor physical anomalies in the AeSOP first-episode psychosis study. Br J Psychiatry 2006;189:221–8. Derecki NC, Cardani AN, Yang CH, Quinnies KM, Crihfield A, Lynch KR, et al. Regulation of learning and memory by meningeal immunity: a key role for IL-4. J Exp Med 2010;207:1067–80. Dickerson F, Boronow J, Stallings C, Origoni A, Yolken R. Toxoplasma gondii in individuals with schizophrenia: association with clinical and demographic factors and with mortality. Schizophr Bull 2007;33:737–40. Dickerson F, Stallings C, Origoni A, Vaughan C, Khushalani S, Yolken R. Additive effects of elevated C-reactive protein and exposure to herpes simplex virus type 1 on cognitive impairment in individuals with schizophrenia. Schizophr Res 2012;134:83–8. Dietz D, Vogel M, Rubin S, Moran T, Carbone K, Pletnikov M. Developmental alterations in serotoninergic neurotransmission in Borna disease virus (BDV)-infected rats: a multidisciplinary analysis. J Neurovirol 2004;10:267–77. Doorduin J, de Vries EF, Willemsen AT, de Groot JC, Dierckx RA, Klein HC. Neuroinflammation in schizophrenia-related psychosis: a PET study. J Nucl Med 2009;50:1801–7. Drexhage RC, Knijff EM, Padmos RC, Heul-Nieuwenhuijzen L, Beumer W, Versnel MA, et al. The mononuclear phagocyte system and its cytokine inflammatory networks in schizophrenia and bipolar disorder. Expert Rev Neurother 2010a;10:59–76. Drexhage RC, van der Heul-Nieuwenhuijsen L, Padmos RC, van Beveren N, Cohen D, Versnel MA, et al. Inflammatory gene expression in monocytes of patients with schizophrenia: overlap and difference with bipolar disorder. A study in naturalistically treated patients. Int J Neuropsychopharmacol 2010b;13:1369–81. Dunne DW, Cooke A. A worm's eye view of the immune system: consequences for evolution of human autoimmune disease. Nat Rev Immunol 2005;5:420–6. Durrwald R, Kolodziejek J, Muluneh A, Herzog S, Nowotny N. Epidemiological pattern of classical Borna disease and regional genetic clustering of Borna disease viruses point towards the existence of to-date unknown endemic reservoir host populations. Microbes Infect 2006;8:917–29. Durrwald R, Kolodziejek J, Herzog S, Nowotny N. Meta-analysis of putative human bornavirus sequences fails to provide evidence implicating Borna disease virus in mental illness. Rev Med Virol 2007;17:181–203. Dworkin J, Shah IM. Exit from dormancy in microbial organisms. Nat Rev Microbiol 2010;8:890–6. Eack SM, Hogarty GE, Cho RY, Prasad KM, Greenwald DP, Hogarty SS, et al. Neuroprotective effects of cognitive enhancement therapy against gray matter loss in early schizophrenia: results from a 2-year randomized controlled trial. Arch Gen Psychiatry 2010;67:674–82. Ehrenreich H, Bartels C, Sargin D, Stawicki S, Krampe H. Recombinant human erythropoietin in the treatment of human brain disease: focus on cognition. J Ren Nutr 2008;18:146–53. Ekdahl CT, Kokaia Z, Lindvall O. Brain inflammation and adult neurogenesis: the dual role of microglia. Neuroscience 2009;158:1021–9. Engelhardt B, Sorokin L. The blood–brain and the blood–cerebrospinal fluid barriers: function and dysfunction. Semin Immunopathol 2009;31:497–511. Fallon BA, Lipkin RB, Corbera KM, Yu S, Nobler MS, Keilp JG, et al. Regional cerebral blood flow and metabolic rate in persistent Lyme encephalopathy. Arch Gen Psychiatry 2009;66:554–63. Fatemi SH, Folsom TD. The neurodevelopmental hypothesis of schizophrenia, revisited. Schizophr Bull 2009;35:528–48. Fatemi SH, Folsom TD, Rooney RJ, Mori S, Kornfield TE, Reutiman TJ, et al. The viral theory of schizophrenia revisited: abnormal placental gene expression and structural changes with lack of evidence for H1N1 viral presence in placentae of infected mice or brains of exposed offspring. Neuropharmacology 2012;62:1290–8. Fathman CG, Soares L, Chan SM, Utz PJ. An array of possibilities for the study of autoimmunity. Nature 2005;435:605–11. Feschotte C. Virology: Bornavirus enters the genome. Nature 2010;463:39–40. Feschotte C, Gilbert C. Endogenous viruses: insights into viral evolution and impact on host biology. Nat Rev Genet 2012;13:283–96. Flyckt L, Borg J, Borg K, Ansved T, Edman G, Bjerkenstedt L, et al. Muscle biopsy, macro EMG, and clinical characteristics in patients with schizophrenia. Biol Psychiatry 2000;47:991–9. Foxe JJ, Yeap S, Snyder AC, Kelly SP, Thakore JH, Molholm S. The N1 auditory evoked potential component as an endophenotype for schizophrenia: high-density electrical mapping in clinically unaffected first-degree relatives, first-episode, and chronic schizophrenia patients. Eur Arch Psychiatry Clin Neurosci 2011;261:331–9. Fusar-Poli P, Borgwardt S, Crescini A, Deste G, Kempton MJ, Lawrie S, et al. Neuroanatomy of vulnerability to psychosis: a voxel-based meta-analysis. Neurosci Biobehav Rev 2011a;35:1175–85. Fusar-Poli P, Crossley N, Woolley J, Carletti F, Perez-Iglesias R, Broome M, et al. Gray matter alterations related to P300 abnormalities in subjects at high risk for psychosis: longitudinal MRI–EEG study. Neuroimage 2011b;55:320–8. Fusar-Poli P, Howes OD, Allen P, Broome M, Valli I, Asselin MC, et al. Abnormal prefrontal activation directly related to pre-synaptic striatal dopamine dysfunction in people at clinical high risk for psychosis. Mol Psychiatry 2011c;16:67–75. Fuxe K, Dahlstrom AB, Jonsson G, Marcellino D, Guescini M, Dam M, et al. The discovery of central monoamine neurons gave volume transmission to the wired brain. Prog Neurobiol 2010;90:82-100. Garver DL, Tamas RL, Holcomb JA. Elevated interleukin-6 in the cerebrospinal fluid of a previously delineated schizophrenia subtype. Neuropsychopharmacology 2003;28: 1515–20. Garver DL, Holcomb JA, Christensen JD. Compromised myelin integrity during psychosis with repair during remission in drug-responding schizophrenia. Int J Neuropsychopharmacol 2008;11:49–61. Gattaz WF, Abrahao AL, Foccacia R. Childhood meningitis, brain maturation and the risk of psychosis. Eur Arch Psychiatry Clin Neurosci 2004;254:23–6. Gawryluk JW, Wang JF, Andreazza AC, Shao L, Young LT. Decreased levels of glutathione, the major brain antioxidant, in post-mortem prefrontal cortex from patients with psychiatric disorders. Int J Neuropsychopharmacol 2011;14:123–30. Glaser CA, Honarmand S, Anderson LJ, Schnurr DP, Forghani B, Cossen CK, et al. Beyond viruses: clinical profiles and etiologies associated with encephalitis. Clin Infect Dis 2006;43:1565–77. Gleeson M, Bishop NC, Stensel DJ, Lindley MR, Mastana SS, Nimmo MA. The anti-inflammatory effects of exercise: mechanisms and implications for the prevention and treatment of disease. Nat Rev Immunol 2011;11:607–15. Glezer I, Simard AR, Rivest S. Neuroprotective role of the innate immune system by microglia. Neuroscience 2007;147:867–83. Goldmann J, Kwidzinski E, et al. T cells traffic from brain to cervical lymph nodes via the cribroid plate and the nasal mucosa. J Leukoc Biol 2006;80(4):797–801. Goldmann J, Kwidzinski E, Brandt C, Mahlo J, Richter D, Bechmann I. T cells traffic from brain to cervical lymph nodes via the cribroid plate and the nasal mucosa. J Leukoc Biol 2006;80:797–801. Gorczynski LY, Gorczynski CP, Terzioglu T, Gorczynski R. Pre- and postnatal influences of neurohormonal triggering and behaviour on the immune system of offspring. Adv Neuroimmune Biol 2011;1:39–51. Grace PM, Rolan PE, Hutchinson MR. Peripheral immune contributions to the maintenance of central glial activation underlying neuropathic pain. Brain Behav Immun 2011;25(7):1322–32. Graham AL, Hayward AD, Watt KA, Pilkington JG, Pemberton JM, Nussey DH. Fitness correlates of heritable variation in antibody responsiveness in a wild mammal. Science 2010;330:662–5. Grassiot B, Desgranges B, Eustache F, Defer G. Quantification and clinical relevance of brain atrophy in multiple sclerosis: a review. J Neurol 2009;256:1397–412. Graus F, Boronat A, Xifro X, Boix M, Svigelj V, Garcia A, et al. The expanding clinical profile of anti-AMPA receptor encephalitis. Neurology 2010;74:857–9. Gross G, Huber G. Somatically based psychoses and the problem of symptomatic schizophrenia. Neurol Psychiatry Brain Res 2007;14:131–42. Gupta S, Soellinger M, Boesiger P, Poulikakos D, Kurtcuoglu V. Three-dimensional computational modeling of subject-specific cerebrospinal fluid flow in the subarachnoid space. J Biomech Eng 2009;131:021010. Gupta S, Soellinger M, Grzybowski DM, Boesiger P, Biddiscombe J, Poulikakos D, et al. Cerebrospinal fluid dynamics in the human cranial subarachnoid space: an overlooked mediator of cerebral disease. I. Computational model. J R Soc Interface 2010;7:1195–204. Hanson DR, Gottesman II. Theories of schizophrenia: a genetic–inflammatory–vascular synthesis. BMC Med Genet 2005;6:7. Harrison PJ, GRoberts GW. The neuropathology of schizophrenia — progress and interpretation. New York: Oxford University Press Inc.; 2000. Hellweg R, von Richthofen S, Anders D, Baethge C, Ropke S, Hartung HD, et al. The time course of nerve growth factor content in different neuropsychiatric diseases — a unifying hypothesis. J Neural Transm 1998;105:871–903. Herden C, Herzog S, Richt JA, Nesseler A, Christ M, Failing K, et al. Distribution of Borna disease virus in the brain of rats infected with an obesity-inducing virus strain. Brain Pathol 2000;10:39–48. Herden C, Schluesener HJ, Richt JA. Expression of allograft inflammatory factor-1 and haeme oxygenase-1 in brains of rats infected with the neurotropic Borna disease virus. Neuropathol Appl Neurobiol 2005;31:512–21. Herrington JD, Schultz RT. Neuroimaging of autism spectrum disorders. In: Shenton ME, Turetsky BI, editors. Understanding neuropsychiatric disorders — insights from neuroimaging. New York: Cambridge University Press; 2011. p. 517. Herzog S, Rott R. Replication of Borna disease virus in cell cultures. Med Microbiol Immunol 1980;168:153–8. Herzog S, Frese K, Rott R. Studies on the genetic control of resistance of black hooded rats to Borna disease. J Gen Virol 1991;72(Pt 3):535–40. Herzog S, Pfeuffer I, Haberzettl K, Feldmann H, Frese K, Bechter K, et al. Molecular characterization of Borna disease virus from naturally infected animals and possible links to human disorders. Arch Virol Suppl 1997;13:183–90. Herzog S, Herden C, Frese K, Lange-Herbst H, Grabner A. Diagnostik der BDV-Infektion beim Pferd: Widersprüche zwischen Intra-vitam- und Post-mortem-Untersuchungen. Pferdeheilkunde 2008;24:766–74. Heuser M, Thomann PA, Essig M, Bachmann S, Schroder J. Neurological signs and morphological cerebral changes in schizophrenia: an analysis of NSS subscales in patients with first episode psychosis. Psychiatry Res 2011;192:69–76. Hobbs JA. Detection of adeno-associated virus 2 and parvovirus B19 in the human dorsolateral prefrontal cortex. J Neurovirol 2006;12:190–9. Holmes E, Tsang TM, Huang JT, Leweke FM, Koethe D, Gerth CW, et al. Metabolic profiling of CSF: evidence that early intervention may impact on disease progression and outcome in schizophrenia. PLoS Med 2006;3:e327. Holtzheimer PE, Mayberg HS. Neuroimaging of mood disorders: commentary. In: Shenton ME, Turetsky BI, editors. Understanding neuropsychiatric disorders — insights from neuroimaging. New York: Cambridge University Press; 2011. p. 197. Horie M, Honda T, Suzuki Y, Kobayashi Y, Daito T, Oshida T, et al. Endogenous non-retroviral RNA virus elements in mammalian genomes. Nature 2010;463:84–7. Hornig M, Briese T, Licinio J, Khabbaz RF, Altshuler LL, Potkin SG, et al. Absence of evidence for bornavirus infection in schizophrenia, bipolar disorder and major depressive disorder. Mol Psychiatry 2012;17(5):486–93. Houtchens MK, Benedict RH, Killiany R, Sharma J, Jaisani Z, Singh B, et al. Thalamic atrophy and cognition in multiple sclerosis. Neurology 2007;69:1213–23. Huang JT, Leweke FM, Oxley D, Wang L, Harris N, Koethe D, et al. Disease biomarkers in cerebrospinal fluid of patients with first-onset psychosis. PLoS Med 2006;3:e428. Huang JT, Leweke FM, Tsang TM, Koethe D, Kranaster L, Gerth CW, et al. CSF metabolic and proteomic profiles in patients prodromal for psychosis. PLoS One 2007;2:e756. K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 Huber G, Gross G, Schuttler R. Schizophrenia. Clinical course and social psychiatric long term examinations of schizophrenic patients hospitalized in Bonn from 1945–1959. Monogr Gesamtgeb Psychiatr Psychiatry Ser 1979;21:1-399. Insel TR. Rethinking schizophrenia. Nature 2010;468:187–93. Irani SR, Vincent A. Autoimmune encephalitis — new awareness, challenging questions. Discov Med 2011;11:449–58. Irani SR, Bera K, Waters P, Zuliani L, Maxwell S, Zandi MS, et al. N-methyl-D-aspartate antibody encephalitis: temporal progression of clinical and paraclinical observations in a predominantly non-paraneoplastic disorder of both sexes. Brain 2010;133:1655–67. Jefferies C, Wynne C, Higgs R. Antiviral TRIMs: friend or foe in autoimmune and autoinflammatory disease? Nat Rev Immunol 2011;11:617–25. Kaminski M, Bechmann I, et al. Migration of monocytes after intracerebral injection at entorhinal cortex lesion site. J Leukoc Biol 2012. Kamitani W, Ono E, Yoshino S, Kobayashi T, Taharaguchi S, Lee BJ, et al. Glial expression of Borna disease virus phosphoprotein induces behavioral and neurological abnormalities in transgenic mice. Proc Natl Acad Sci U S A 2003;100:8969–74. Kang HJ, Kawasawa YI, Cheng F, Zhu Y, Xu X, Li M, et al. Spatio-temporal transcriptome of the human brain. Nature 2011;478:483–9. Keizman D, Rogowski O, et al. Low-grade systemic inflammation in patients with amyotrophic lateral sclerosis. Acta Neurol Scand 2009;119(6):383–9. Kerschensteiner M, Meinl E, Hohlfeld R. Neuro-immune crosstalk in CNS diseases. Neuroscience 2009;158:1122–32. Kirkbride JB, Scoriels L. Review of the 6th symposium for the search for the causes of schizophrenia, Sao Paulo, Brazil, 3–6 February 2009. Eur Arch Psychiatry Clin Neurosci 2009;259:505–9. Kleines M, Schiefer J, Stienen A, Blaum M, Ritter K, Hausler M. Expanding the spectrum of neurological disease associated with Epstein–Barr virus activity. Eur J Clin Microbiol Infect Dis 2011;30:1561–9. Klosterkötter J. Early detection of schizophrenia. Neurol Psychiatry Brain Res 2011;17: 1-10. Klosterkotter J, Gross G, Huber G. The concept of process activity in idiopathic psychoses. Nervenarzt 1989;60:740–4. Kochunov P, Glahn DC, Nichols TE, Winkler AM, Hong EL, Holcomb HH, et al. Genetic analysis of cortical thickness and fractional anisotropy of water diffusion in the brain. Front Neurosci 2011;5:120. Koponen H, Rantakallio P, Veijola J, Jones P, Jokelainen J, Isohanni M. Childhood central nervous system infections and risk for schizophrenia. Eur Arch Psychiatry Clin Neurosci 2004;254:9-13. Koster-Patzlaff C, Hosseini SM, Reuss B. Persistent Borna disease virus infection changes expression and function of astroglial gap junctions in vivo and in vitro. Brain Res 2007;1184:316–32. Kreutzberg GW. Microglia: a sensor for pathological events in the CNS. Trends Neurosci 1996;19:312–8. Krishnamoorthy G, Saxena A, Mars LT, Domingues HS, Mentele R, Ben-Nun A, et al. Myelin-specific T cells also recognize neuronal autoantigen in a transgenic mouse model of multiple sclerosis. Nat Med 2009;15:626–32. Kurian MA, Gissen P, Smith M, Heales Jr S, Clayton PT. The monoamine neurotransmitter disorders: an expanding range of neurological syndromes. Lancet Neurol 2011;10:721–33. Lang UE, Jockers-Scherubl MC, Hellweg R. State of the art of the neurotrophin hypothesis in psychiatric disorders: implications and limitations. J Neural Transm 2004;111:387–411. Lawn SD, Zumla AI. Tuberculosis. Lancet 2011;378:57–72. Leboyer M, Tamouza R, Charron D, Faucard R, Perron H. Human endogenous retrovirus type W (HERV-W) in schizophrenia: a new avenue of research at the gene–environment interface. World J Biol Psychiatry 2011. Lederbogen F, Kirsch P, Haddad L, Streit F, Tost H, Schuch P, et al. City living and urban upbringing affect neural social stress processing in humans. Nature 2011;474:498–501. Lehtinen MK, Zappaterra MW, Chen X, Yang YJ, Hill AD, Lun M, et al. The cerebrospinal fluid provides a proliferative niche for neural progenitor cells. Neuron 2011;69:893–905. Lencer R, Keedy SK, Reilly JL, McDonough BE, Harris MS, Sprenger A, et al. Altered transfer of visual motion information to parietal association cortex in untreated first-episode psychosis: implications for pursuit eye tracking. Psychiatry Res 2011;194:30–8. Lennox BR, Coles AJ, Vincent A. Antibody-mediated encephalitis: a treatable cause of schizophrenia. Br J Psychiatry 2012;200:92–4. Leweke FM, Gerth CW, Koethe D, Klosterkotter J, Ruslanova I, Krivogorsky B, et al. Antibodies to infectious agents in individuals with recent onset schizophrenia. Eur Arch Psychiatry Clin Neurosci 2004;254:4–8. Li F, Nellaker C, Yolken RH, Karlsson H. A systematic evaluation of expression of HERV-W elements; influence of genomic context, viral structure and orientation. BMC Genomics 2011;12:22. Licinio J. Potential diagnostic markers for postpartum depression point out to altered immune signaling. Mol Psychiatry 2010;15:1. Licinio J, Wong ML. The role of inflammatory mediators in the biology of major depression: central nervous system cytokines modulate the biological substrate of depressive symptoms, regulate stress-responsive systems, and contribute to neurotoxicity and neuroprotection. Mol Psychiatry 1999;4:317–27. Lieb K, Hufert FT, Bechter K, Bauer J, Kornhuber J. Depression, Borna disease, and amantadine. Lancet 1997;349:958. Lipkin WI, Briese T, Hornig M. Borna disease virus — fact and fantasy. Virus Res 2011;162:162–72. Locatelli G, Wortge S, Buch T, Ingold B, Frommer F, Sobottka B, et al. Primary oligodendrocyte death does not elicit anti-CNS immunity. Nat Neurosci 2012;15(4): 543–50. Lucchinetti CF, Popescu BF, Bunyan RF, Moll NM, Roemer SF, Lassmann H, et al. Inflammatory cortical demyelination in early multiple sclerosis. N Engl J Med 2011;365:2188–97. 89 Lynch MA, Mills KH. Immunology meets neuroscience — opportunities for immune intervention in neurodegenerative diseases. Brain Behav Immun 2012;26(1):1-10. MacKenzie-Graham A, Tinsley MR, Shah KP, Aguilar C, Strickland LV, Boline J, et al. Cerebellar cortical atrophy in experimental autoimmune encephalomyelitis. Neuroimage 2006;32:1016–23. Maes M. Cytokines in schizophrenia. Biol Psychiatry 1997;42:308–9. Marques F, Sousa JC, Coppola G, Geschwind DH, Sousa N, Palha JA, et al. The choroid plexus response to a repeated peripheral inflammatory stimulus. BMC Neurosci 2009;10:135. Marshall BJ. Campylobacter pylori: its link to gastritis and peptic ulcer disease. Rev Infect Dis 1990;12(Suppl. 1):S87–93. Martins-de-Souza D, Lebar M, Turck CW. Proteome analyses of cultured astrocytes treated with MK-801 and clozapine: similarities with schizophrenia. Eur Arch Psychiatry Clin Neurosci 2011;261:217–28. Matzinger P, Kamala T. Tissue-based class control: the other side of tolerance. Nat Rev Immunol 2011;11:221–30. Maxeiner HG, Rojewski MT, Schmitt A, Tumani H, Bechter K, Schmitt M. Flow cytometric analysis of T cell subsets in paired samples of cerebrospinal fluid and peripheral blood from patients with neurological and psychiatric disorders. Brain Behav Immun 2009;23:134–42. McGeer PL, McGeer EG. The inflammatory response system of brain: implications for therapy of Alzheimer and other neurodegenerative diseases. Brain Res Brain Res Rev 1995;21:195–218. McLeod JG, Hammond SR, Kurtzke JF. Migration and multiple sclerosis in immigrants to Australia from United Kingdom and Ireland: a reassessment. I. Risk of MS by age at immigration. J Neurol 2011;258:1140–9. Medzhitov R. Origin and physiological roles of inflammation. Nature 2008;454:428–35. Meinl E, Krumbholz M, Hohlfeld R. B lineage cells in the inflammatory central nervous system environment: migration, maintenance, local antibody production, and therapeutic modulation. Ann Neurol 2006;59:880–92. Meltzer HY, Crayton JW. Subterminal motor nerve abnormalities in psychotic patients. Nature 1974;249:373–5. Menninger KA. Influenza and schizophrenia. An analysis of post-influenzal “dementia precox,” as of 1918, and five years later further studies of the psychiatric aspects of influenza. 1926. Am J Psychiatry 1994;151:182–7. Meyer U. Anti-inflammatory signaling in schizophrenia. Brain Behav Immun 2011;25(8):1507–18. Meyer U, Nyffeler M, Engler A, Urwyler A, Schedlowski M, Knuesel I, et al. The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. J Neurosci 2006;26:4752–62. Meyer JM, McEvoy JP, Davis VG, Goff DC, Nasrallah HA, Davis SM, et al. Inflammatory markers in schizophrenia: comparing antipsychotic effects in phase 1 of the clinical antipsychotic trials of intervention effectiveness study. Biol Psychiatry 2009;66: 1013–22. Miller J, Drost DJ, Jensen E, Manchanda R, Northcott S, Neufeld RW, et al. Progressive membrane phospholipid changes in first episode schizophrenia with high field magnetic resonance spectroscopy. Psychiatry Res 2012;201:25–33. Mills KH. TLR-dependent T cell activation in autoimmunity. Nat Rev Immunol 2011;11: 807–22. Minshew NJ. Neuroimaging of developmental: commentary. In: Shenton ME, Turetsky BI, editors. Understanding neuropsychiatric disorders — insights from neuroimaging. New York: Cambridge University Press; 2011. p. 555. Mulert C, Kirsch V, Pascual-Marqui R, McCarley RW, Spencer KM. Long-range synchrony of gamma oscillations and auditory hallucination symptoms in schizophrenia. Int J Psychophysiol 2011a;79:55–63. Mulert C, Kirsch V, Whitford TJ, Alvarado J, Pelavin P, McCarley RW, et al. Hearing voices: a role of interhemispheric auditory connectivity? World J Biol Psychiatry 2011b. Muller N, Schwarz MJ. Immune system and schizophrenia. Curr Immunol Rev 2010;6: 213–20. Musgrave T, Benson C, Wong G, Browne I, Tenorio G, Rauw G, et al. The MAO inhibitor phenelzine improves functional outcomes in mice with experimental autoimmune encephalomyelitis (EAE). Brain Behav Immun 2011;25:1677–88. Mutnal MB, Hu S, Little MR, Lokensgard JR. Memory T cells persisting in the brain following MCMV infection induce long-term microglial activation via interferon-gamma. J Neurovirol 2011;17:424–37. Myint AM, Kim YK. Cytokine–serotonin interaction through IDO: a neurodegeneration hypothesis of depression. Med Hypotheses 2003;61:519–25. Myint AM, Schwarz MJ, Verkerk R, Mueller HH, Zach J, Scharpe S, et al. Reversal of imbalance between kynurenic acid and 3-hydroxykynurenine by antipsychotics in medication-naive and medication-free schizophrenic patients. Brain Behav Immun 2011;25(8):1576–81. Najmabadi H, Hu H, Garshasbi M, Zemojtel T, Abedini SS, Chen W, et al. Deep sequencing reveals 50 novel genes for recessive cognitive disorders. Nature 2011;478:57–63. Nathan C. Points of control in inflammation. Nature 2002;420(6917):846–52. Nave KA. Myelination and support of axonal integrity by glia. Nature 2010;468:244–52. Niebuhr DW, Millikan AM, Cowan DN, Yolken R, Li Y, Weber NS. Selected infectious agents and risk of schizophrenia among U.S. military personnel. Am J Psychiatry 2008;165:99-106. Nielsen PR, Laursen TM, Mortensen PB. Association between parental hospital-treated infection and the risk of schizophrenia in adolescence and early adulthood. Schizophr Bull 2011. Nieratschker V, Nothen MM, Rietschel M. New genetic findings in schizophrenia: is there still room for the dopamine hypothesis of schizophrenia? Front Behav Neurosci 2010;4:23. Nikisch G, Baumann P, Liu T, Mathe AA. Quetiapine affects neuropeptide Y and corticotropin-releasing hormone in cerebrospinal fluid from schizophrenia patients: 90 K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 relationship to depression and anxiety symptoms and to treatment response. Int J Neuropsychopharmacol 2011:1-11. Nord M, Farde L. Antipsychotic occupancy of dopamine receptors in schizophrenia. CNS Neurosci Ther 2011;17:97-103. O'Callaghan JP, Sriram K, Miller DB. Defining “neuroinflammation”. Ann N Y Acad Sci 2008;1139:318–30. O'Connor SM, Taylor CE, Hughes JM. Emerging infectious determinants of chronic diseases. Emerg Infect Dis 2006;12:1051–7. Oldstone MB. Molecular anatomy of viral disease. The George Cotzias memorial lecture 1986. Neurology 1987;37:453–60. Oldstone MBA. Viruses can cause disease in the absence of morphologic evidence of cell injury: pathology in the absence of cell lysis — implication for pathologist's future study of disease. In: Cancilla PA, Vogel F, editors. Neuropathology. New York: Williams; 1990. p. 123–9. Oleszak EL, Chang JR, Friedman H, Katsetos CD, Platsoucas CD. Theiler's virus infection: a model for multiple sclerosis. Clin Microbiol Rev 2004;17:174–207. Oleszak EL, Zhang X, Lin WL, Lu S, Chang JR, Tsygankov A, et al. Immunopathology of multiple sclerosis and psychoses. In Vivo 2007;21:945. Osborn AG, Salzman KL, Barkovich AJ. Diagnostic imaging: brain. Lippincott Williams & Wilkins; 2010. Ovanesov MV, Ayhan Y, Wolbert C, Moldovan K, Sauder C, Pletnikov MV. Astrocytes play a key role in activation of microglia by persistent Borna disease virus infection. J Neuroinflammation 2008;5. art. no. 50. Owen MJ, O'Donovan MC, Thapar A, Craddock N. Neurodevelopmental hypothesis of schizophrenia. Br J Psychiatry 2011;198:173–5. Pantelis C, Velakoulis D, Wood SJ, Yucel M, Yung AR, Phillips LJ, et al. Neuroimaging and emerging psychotic disorders: the Melbourne ultra-high risk studies. Int Rev Psychiatry 2007;19:371–81. Peerbooms OL, van Os J, Drukker M, Kenis G, Hoogveld L, de Hert M, et al. Meta-analysis of MTHFR gene variants in schizophrenia, bipolar disorder and unipolar depressive disorder: evidence for a common genetic vulnerability? Brain Behav Immun 2011;25:1530–43. Perron H, Lang A. The human endogenous retrovirus link between genes and environment in multiple sclerosis and in multifactorial diseases associating neuroinflammation. Clin Rev Allergy Immunol 2010;39:51–61. Perron H, Mekaoui L, Bernard C, Veas F, Stefas I, Leboyer M. Endogenous retrovirus type W GAG and envelope protein antigenemia in serum of schizophrenic patients. Biol Psychiatry 2008;64:1019–23. Picard D, Janela B, Descamps V, D'Incan M, Courville P, Jacquot S, et al. Drug reaction with eosinophilia and systemic symptoms (DRESS): a multiorgan antiviral T cell response. Sci Transl Med 2010;2:46ra62. Piontkewitz Y, Assaf Y, Weiner I. Clozapine administration in adolescence prevents postpubertal emergence of brain structural pathology in an animal model of schizophrenia. Biol Psychiatry 2009;66:1038–46. Planz O, Bechter K, Schwemmle M. Human Borna disease virus infection. In: Carbone K, editor. Borna disease virus — role in neurobehavioral disease. Washington: ASM Press; 2002. p. 179–225. Pletnikov M, Gonzalez-Dunia D, Stitz L. Experimental infection: pathogenesis of neurobehavioral disease. In: Carbone K, editor. Borna disease virus and its role in neurobehavioral disease. Washington, DC: ASM Press; 2002. p. 125–78. Pletnikov MV, Ayhan Y, Nikolskaia O, Xu Y, Ovanesov MV, Huang H, et al. Inducible expression of mutant human DISC1 in mice is associated with brain and behavioral abnormalities reminiscent of schizophrenia. Mol Psychiatry 2008;13(173–86): 115. Prandovszky E, Gaskell E, Martin H, Dubey JP, Webster JP, McConkey GA. The neurotropic parasite Toxoplasma gondii increases dopamine metabolism. PLoS One 2011;6:e23866. Prasad KM, Shirts BH, Yolken RH, Keshavan MS, Nimgaonkar VL. Brain morphological changes associated with exposure to HSV1 in first-episode schizophrenia. Mol Psychiatry 2007;12(105–13):1. Prasad KM, Eack SM, Goradia D, Pancholi KM, Keshavan MS, Yolken RH, et al. Progressive gray matter loss and changes in cognitive functioning associated with exposure to herpes simplex virus 1 in schizophrenia: a longitudinal study. Am J Psychiatry 2011;168:822–30. Pruss H, Dalmau J, Harms L, Holtje M, Ahnert-Hilger G, Borowski K, et al. Retrospective analysis of NMDA receptor antibodies in encephalitis of unknown origin. Neurology 2010;75:1735–9. Pugliese A, Beltramo T, Torre D, Roccatello D. Parvovirus B19 and immune disorders. Cell Biochem Funct 2007;25:639–41. Raivich G, Jones LL, Werner A, Bluthmann H, Doetschmann T, Kreutzberg GW. Molecular signals for glial activation: pro- and anti-inflammatory cytokines in the injured brain. Acta Neurochir Suppl 1999;73:21–30. Ransohoff RM. Immunology: in the beginning. Nature 2009;462:41–2. Redzic Z. Molecular biology of the blood-brain and the blood-cerebrospinal fluid barriers: similarities and differences. Fluids Barriers CNS 2011;8(1):3. Reiber H, Peter JB. Cerebrospinal fluid analysis: disease-related data patterns and evaluation programs. J Neurol Sci 2001;184:101–22. Ribbe K, Ackermann V, Schwitulla J, Begemann M, Papiol S, Grube S, et al. Prediction of the risk of comorbid alcoholism in schizophrenia by interaction of common genetic variants in the corticotropin-releasing factor system. Arch Gen Psychiatry 2011;68: 1247–56. Richt JA, Alexander RC, Herzog S, Hooper DC, Kean R, Spitsin S, et al. Failure to detect Borna disease virus infection in peripheral blood leukocytes from humans with psychiatric disorders. J Neurovirol 1997a;3:174–8. Richt JA, Pfeuffer I, Christ M, Frese K, Bechter K, Herzog S. Borna disease virus infection in animals and humans. Emerg Infect Dis 1997b;3:343–52. Riecher-Rossler A, Pflueger MO, Aston J, Borgwardt SJ, Brewer WJ, Gschwandtner U, et al. Efficacy of using cognitive status in predicting psychosis: a 7-year follow-up. Biol Psychiatry 2009;66:1023–30. Rioux JD, Abbas AK. Paths to understanding the genetic basis of autoimmune disease. Nature 2005;435:584–9. Rocca MA, Mondria T, Valsasina P, Sormani MP, Flach ZH, Te Boekhorst PA, et al. A three-year study of brain atrophy after autologous hematopoietic stem cell transplantation in rapidly evolving secondary progressive multiple sclerosis. AJNR Am J Neuroradiol 2007;28:1659–61. Rolls ET, Loh M, Deco G, Winterer G. Computational models of schizophrenia and dopamine modulation in the prefrontal cortex. Nat Rev Neurosci 2008;9:696–709. Rott R, Herzog S, Bechter K, Frese K. Borna disease, a possible hazard for man? Arch Virol 1991;118:143–9. Rott O, Herzog S, Cash E. T cell memory specific for self and non-self antigens in rats persistently infected with Borna disease virus. Clin Exp Immunol 1993;93:370–6. Rujescu D. Are we going to end up with many distinct genomic syndromes in psychiatry? Eur Arch Psychiatry Clin Neurosci 2011;261:145–6. Saijo K, Glass CK. Microglial cell origin and phenotypes in health and disease. Nat Rev Immunol 2011;11:775–87. Santos A, Meyer-Lindenberg A. Neuroimaging of Williams–Beuren syndrome. In: Shenton ME, Turetsky BI, editors. Understanding neuropsychiatric disorders — insights from neuroimaging. New York: Cambridge University Press; 2011. p. 537. Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 2011;476:214–9. Saxton KB, John-Henderson N, Reid MW, Francis DD. The social environment and IL-6 in rats and humans. Brain Behav Immun 2011;25:1617–25. Schmitt A, Hasan A, Gruber O, Falkai P. Schizophrenia as a disorder of disconnectivity. Eur Arch Psychiatry Clin Neurosci 2011a;261(Suppl. 2):150–4. Schmitt M, Neubauer A, Greiner J, Xu X, Barth TF, Bechter K. Spreading of acute myeloid leukemia cells by trafficking along the peripheral outflow pathway of cerebrospinal fluid. Anticancer Res 2011b;31:2343–5. Schwartz M, Kipnis J. A conceptual revolution in the relationships between the brain and immunity. Brain Behav Immun 2011;25:817–9. Schwartz M, Shechter R. Protective autoimmunity functions by intracranial immunosurveillance to support the mind: the missing link between health and disease. Mol Psychiatry 2010;15:342–54. Schwarz E, Guest PC, Rahmoune H, Harris LW, Wang L, Leweke FM, et al. Identification of a biological signature for schizophrenia in serum. Mol Psychiatry 2012;17(5):494–502. Schwemmle M. Borna disease virus infection in psychiatric patients: are we on the right track? Lancet Infect Dis 2001;1:46–52. Schwemmle M, Heimrich B. Viral interference with neuronal integrity: what can we learn from the Borna disease virus? Cell Tissue Res 2011;344:13–6. Schwemmle M, Jehle C, Formella S, Staeheli P. Sequence similarities between human bornavirus isolates and laboratory strains question human origin. Lancet 1999;354:1973–4. Shenton ME, Turetsky BI. Understanding neuropsychiatric disorders — insights from neuroimaging. New York: Cambridge University Press; 2011. Simon AE, Velthorst E, Nieman DH, Linszen D, Umbricht D, de Haan L. Ultra high-risk state for psychosis and non-transition: a systematic review. Schizophr Res 2011;132(1):8-17. Skulina C, Schmidt S, Dornmair K, Babbe H, Roers A, Rajewsky K, et al. Multiple sclerosis: brain-infiltrating CD8+ T cells persist as clonal expansions in the cerebrospinal fluid and blood. Proc Natl Acad Sci U S A 2004;101:2428–33. Smiley JF, Rosoklija G, Mancevski B, Pergolizzi D, Figarsky K, Bleiwas C, et al. Hemispheric comparisons of neuron density in the planum temporale of schizophrenia and nonpsychiatric brains. Psychiatry Res 2011;192:1-11. Sprankel H, Richarz K, Ludwig H, Rott R. Behavior alterations in tree shrews (Tupaia glis, Diard 1820) induced by Borna disease virus. Med Microbiol Immunol 1978;165: 1-18. St Clair D, Xu M, Wang P, Yu Y, Fang Y, Zhang F, et al. Rates of adult schizophrenia following prenatal exposure to the Chinese famine of 1959–1961. JAMA 2005;294:557–62. Stanek G, Wormser GP, Gray J, Strle F. Lyme borreliosis. Lancet 2012;379(9814): 461–73. Stanta JL, Saldova R, Struwe WB, Byrne JC, Leweke FM, Rothermund M, et al. Identification of N-glycosylation changes in the CSF and serum in patients with schizophrenia. J Proteome Res 2010;9:4476–89. Stefansson H, Ophoff RA, Steinberg S, Andreassen OA, Cichon S, Rujescu D, et al. Common variants conferring risk of schizophrenia. Nature 2009;460:744–7. Steiner J, Bielau H, Brisch R, Danos P, Ullrich O, Mawrin C, et al. Immunological aspects in the neurobiology of suicide: elevated microglial density in schizophrenia and depression is associated with suicide. J Psychiatr Res 2008;42:151–7. Steiner J, Bogerts B, Sarnyai Z, Walter M, Gos T, Bernstein HG, et al. Bridging the gap between the immune and glutamate hypotheses of schizophrenia and major depression: potential role of glial NMDA receptor modulators and impaired blood–brain barrier integrity. World J Biol Psychiatry 2011. Strik C, Klose U, Erb M, Strik H, Grodd W. Intracranial oscillations of cerebrospinal fluid and blood flows: analysis with magnetic resonance imaging. J Magn Reson Imaging 2002;15:251–8. Sun D, Stuart GW, Jenkinson M, Wood SJ, McGorry PD, Velakoulis D, et al. Brain surface contraction mapped in first-episode schizophrenia: a longitudinal magnetic resonance imaging study. Mol Psychiatry 2009;14:976–86. Susser ES, Lin SP. Schizophrenia after prenatal exposure to the Dutch Hunger Winter of 1944–1945. Arch Gen Psychiatry 1992;49:983–8. Tekok-Kilic A, Benedict RH, Weinstock-Guttman B, Dwyer MG, Carone D, Srinivasaraghavan B, et al. Independent contributions of cortical gray matter atrophy and ventricle K. Bechter / Progress in Neuro-Psychopharmacology & Biological Psychiatry 42 (2013) 71–91 enlargement for predicting neuropsychological impairment in multiple sclerosis. Neuroimage 2007;36:1294–300. Tlaskalova-Hogenova H, Stepankova R, Kozakova H, Hudcovic T, Vannucci L, Tuckova L, et al. The role of gut microbiota (commensal bacteria) and the mucosal barrier in the pathogenesis of inflammatory and autoimmune diseases and cancer: contribution of germ-free and gnotobiotic animal models of human diseases. Cell Mol Immunol 2011;8:110–20. Trushina E, McMurray CT. Oxidative stress and mitochondrial dysfunction in neurodegenerative diseases. Neuroscience 2007;145:1233–48. Uher R. The role of genetic variation in the causation of mental illness: an evolution-informed framework. Mol Psychiatry 2009;14(12):1072–82. Urbach H, Soeder BM, Jeub M, Klockgether T, Meyer B, Bien CG. Serial MRI of limbic encephalitis. Neuroradiology 2006;48:380–6. van Haren NE, Schnack HG, Cahn W, van den Heuvel MP, Lepage C, Collins L, et al. Changes in cortical thickness during the course of illness in schizophrenia. Arch Gen Psychiatry 2011;68:871–80. van Os J, Kenis G, Rutten BP. The environment and schizophrenia. Nature 2010;468:203–12. Verrall L, Burnet PW, Betts JF, Harrison PJ. The neurobiology of D-amino acid oxidase and its involvement in schizophrenia. Mol Psychiatry 2010;15:122–37. Vincent A, Buckley C, Schott JM, Baker I, Dewar BK, Detert N, et al. Potassium channel antibody-associated encephalopathy: a potentially immunotherapy-responsive form of limbic encephalitis. Brain 2004;127:701–12. Vincent A, Bien CG, Irani SR, Waters P. Autoantibodies associated with diseases of the CNS: new developments and future challenges. Lancet Neurol 2011;10:759–72. Voineagu I, Wang X, Johnston P, Lowe JK, Tian Y, Horvath S, et al. Transcriptomic analysis of autistic brain reveals convergent molecular pathology. Nature 2011;474:380–4. Wallin MT, Page WF, Kurtzke JF. Migration and multiple sclerosis in Alaskan military veterans. J Neurol 2009;256:1413–7. Wang H, Yolken RH, Hoekstra PJ, Burger H, Klein HC. Antibodies to infectious agents and the positive symptom dimension of subclinical psychosis: the TRAILS study. Schizophr Res 2011;129:47–51. Werner-Keiss N, Garten W, Richt JA, Porombka D, Algermissen D, Herzog S, et al. Restricted expression of Borna disease virus glycoprotein in brains of experimentally infected Lewis rats. Neuropathol Appl Neurobiol 2008;34:590–602. Westergaard T, Mortensen PB, Pedersen CB, Wohlfahrt J, Melbye M. Exposure to prenatal and childhood infections and the risk of schizophrenia: suggestions from a study of sibship characteristics and influenza prevalence. Arch Gen Psychiatry 1999;56:993–8. White T, Andreasen NC, Nopoulos P, Magnotta V. Gyrification abnormalities in childhood- and adolescent-onset schizophrenia. Biol Psychiatry 2003;54:418–26. 91 White T, Schmidt M, Karatekin C. White matter ‘potholes’ in early-onset schizophrenia: a new approach to evaluate white matter microstructure using diffusion tensor imaging. Psychiatry Res 2009;174:110–5. White T, Su S, Schmidt M, Kao CY, Sapiro G. The development of gyrification in childhood and adolescence. Brain Cogn 2010;72:36–45. White T, Moeller S, Schmidt M, Pardo JV, Olman C. Evidence for intact local connectivity but disrupted regional function in the occipital lobe in children and adolescents with schizophrenia. Hum Brain Mapp 2011a. White T, Schmidt M, Kim DI, Calhoun VD. Disrupted functional brain connectivity during verbal working memory in children and adolescents with schizophrenia. Cereb Cortex 2011b;21:510–8. Whitford TJ, Shenton ME. Structural imaging of schizophrenia. In: Shenton ME, Turetsky BI, editors. Understanding neuropsychiatric disorders. New York: Cambridge University Press; 2011. p. 1-29. Wildemann B, Oschmann P, Reiber H. Laboratory diagnosis in neurology. Stuttgart: Thieme; 2010. Willyard C. Gut reaction. Nature 2011;479:S5–7. Wilson EH, Harris TH, Mrass P, John B, Tait ED, Wu GF, et al. Behavior of parasite-specific effector CD8+ T cells in the brain and visualization of a kinesis-associated system of reticular fibers. Immunity 2009;30:300–11. Wolburg H, Paulus W. Choroid plexus: biology and pathology. Acta Neuropathol 2010;119: 75–88. Wollinsky KH, Hulser PJ, Brinkmeier H, Aulkemeyer P, Bossenecker W, Huber-Hartmann KH, et al. CSF filtration is an effective treatment of Guillain–Barre syndrome: a randomized clinical trial. Neurology 2001;57:774–80. Wood F, Bloor MJ. Borna disease virus: the generation and review of a scientific study. Soc Sci Med 2006;63:1072–83. Yolken RH, Torrey EF. Are some cases of psychosis caused by microbial agents? A review of the evidence. Mol Psychiatry 2008;13:470–9. Yolken RH, Torrey EF, Lieberman JA, Yang S, Dickerson FB. Serological evidence of exposure to herpes simplex virus type 1 is associated with cognitive deficits in the CATIE schizophrenia sample. Schizophr Res 2011;128:61–5. Zandi MS, Irani SR, Lang B, Waters P, Jones PB, McKenna P, et al. Disease-relevant autoantibodies in first episode schizophrenia. J Neurol 2011;258:686–8. zur Hausen H. Cervical carcinoma and human papillomavirus: on the road to preventing a major human cancer. J Natl Cancer Inst 2001;93:252–3.
© Copyright 2026 Paperzz