LEPTIN RESISTANCE REDUCES GROWTH HORMONE SECRETION AND CONTRIBUTES TO THE PATHOGENESIS OF OBESITY By ROBIN LEIGH MARTIN A DISSERTATION PRESENTED TO THE GRADUATE SCHOOL OF THE UNIVERSITY OF FLORIDA IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY UNIVERSITY OF FLORIDA 2000 For Jarod ACKNOWLEDGEMENTS There are many people to whom I wish to express gratitude for standing by me during my efforts to get my Ph.D. First, my family: husband, George Martin; step-son, Joshua Martin; mother, Nikki Picking; father, Tom Picking; brother and sister-in-law, Reed and Kelly Picking, and grandparents, Don and Maxine Picking. They have all been tremendously understanding and supportive, and I greatly appreciate it. I would also like to acknowledge the support I have received from in-laws, aunts, uncles, cousins, nieces, and friends, all of whom have taken the good with the bad. I want to thank my mentor, Dr. William Millard, for his guidance and patience in helping me reach my goal and for his faith in my "independence." Sometimes he had more faith than I did. I would also like to thank the other members of my supervisory committee, Dr. Ralph Dawson, Dr. Maureen Keller-Wood, Dr. James Simpkins, and Dr. Pushpa Kalra, for their valuable advice and for allowing liberal laboratory access and use of equipment. In addition, I thank Dr. Jeff Hughes, Dr. James Simpkins, and Dr. Maureen Keller-Wood for finding extra work for me to do, such as performing surgeries and running assays, when my finances were low. I am sure they benefitted from my help, but not nearly as much as I benefitted from their money. I wish to thank Evelyn Perez, Feng Li, DeeAnn Dugan, and Amanda Crews, students who worked in my laboratory, for patiently allowing me to hone and sharpen my training and supervisory skills. It was fun. Thanks also goes to Dr. Yun-Ju He and Eileen Monck, who were quick to share their extensive technical expertise with me. I am iii grateful to the office staff, Donna Walko, Milena Palenzuela, Gwen Daniels, and especially Theresa Jones, for all of their help in areas about which I know little. I also appreciate the friendships I made with other students with whom I went through graduate school both in my department and from other departments: Dr. Darren Roesch, Dr. Kelly Gridley, Dr. Ming Hu, Dr. Pini Orbach, Dr. Scott Purinton, Tony Smith, and Amanda Crews. I want to give special mention to Dr. Pattie Green, who was a great study partner and friend, especially during our first two years as graduate students. In addition I want to thank Dr. Bruce Jung for all the challenging, and often therapeutic, racquetball games. The most important person I must mention, however, is Dr. Baerbel Eppler. I am thoroughly indebted to BB for her scientific help as well as for her friendship. I would like to thank my friends Tim and Anita Harvey, who very unselfishly provided me with a place to live and who took care of me when I first arrived in Gainesville. I am also grateful to have secured friendships with Dr. Joanna Peris, Dr. LeighAnn Stubley, Dr. Baerbel Eppler, and Theresa Jones while living in Gainesville. Finally, I especially thank all my family and friends in Clearwater, who are awaiting my return. iv TABLE OF CONTENTS Chapter Page ACKNOWLEDGEMENTS iii LIST OF FIGURES ix LIST OF TABLES xi 1 LITERATURE REVIEW 1 Obesity ........................................................................................................................ 1 Obesity Models ........................................................................................................... 3 Theories of Food Intake .............................................................................................. 6 Leptin .......................................................................................................................... 9 Roles of Leptin.......................................................................................................... 12 Regulation of Leptin ................................................................................................. 15 Leptin and Hormonal Interactions ............................................................................ 16 Leptin Receptors ....................................................................................................... 19 Leptin Signaling........................................................................................................ 23 Leptin Resistance ...................................................................................................... 25 Human Leptin Mutations .......................................................................................... 28 Leptin Treatment in Humans and Leptin Gene Therapy .......................................... 29 Growth Hormone ...................................................................................................... 30 Excess of Deficiency of Growth Hormone ............................................................... 35 Growth Hormone and Obesity.................................................................................. 36 Leptin and Growth Hormone .................................................................................... 39 Objectives.................................................................................................................. 41 2 GENERAL METHODS 43 Animals ..................................................................................................................... 43 Diets .......................................................................................................................... 43 Feeding, Pair-feeding, and Body Weight Measurements ......................................... 43 Leptin Challenge Test............................................................................................... 44 Alzet Osmotic Minipumps ........................................................................................ 45 Right Atrial Cannulation........................................................................................... 45 Anesthesia ............................................................................................................. 45 Preparation ............................................................................................................ 45 v Surgery.................................................................................................................. 46 Recovery ............................................................................................................... 46 Blood sampling cages ............................................................................................... 46 Blood Sampling and Tissue Collection..................................................................... 47 Blood Collection from Cannulae .......................................................................... 47 Blood Collection from Tail Vein .......................................................................... 47 Blood Collection via Cardiac Puncture................................................................. 47 Trunk Blood Collection ........................................................................................ 48 Collection of Hypothalamus ................................................................................. 48 Pituitary and Organ Weights................................................................................. 48 Cell Culture............................................................................................................... 48 GH1 Cells.............................................................................................................. 48 Plating Cells .......................................................................................................... 49 Five-Day Time-Course ......................................................................................... 49 Media Experiment................................................................................................. 49 Collecting RNA from GH1 Cells.......................................................................... 50 Radioimmunoassays.................................................................................................. 50 GH Iodination ....................................................................................................... 50 Growth Hormone RIA .......................................................................................... 51 IGF Iodination....................................................................................................... 51 IGF RIA ................................................................................................................ 52 Leptin RIA ............................................................................................................ 52 Insulin RIA............................................................................................................ 53 Glucose and Triglyceride Assays.............................................................................. 54 DNA Assay ............................................................................................................... 54 RNA extraction, RT-PCR, Southern Blot................................................................. 55 Protein Extraction and Western Immunoblotting ..................................................... 57 Protein Extraction ................................................................................................. 57 Micro BCA Protein Assay .................................................................................... 58 Western Immunoblot Analysis for Leptin Receptor ............................................. 59 3 CIRCULATING GROWTH HORMONE LEVELS ARE ELEVATED IN RATS FED A HIGH-FAT DIET 62 Introduction............................................................................................................... 62 Methods..................................................................................................................... 63 Animals ................................................................................................................. 63 Cannulation Surgery, Blood Sampling, and Tissue Collection ............................ 64 Radioimmunoassays.............................................................................................. 65 RT-PCR for Leptin Receptor ................................................................................ 65 Statistics .................................................................................................................... 66 Results ....................................................................................................................... 66 Body Weight ......................................................................................................... 66 Food Intake ........................................................................................................... 66 Leptin .................................................................................................................... 67 Leptin Receptor mRNA ........................................................................................ 67 vi IGF ........................................................................................................................ 67 Growth Hormone Profile ...................................................................................... 67 Discussion ................................................................................................................. 72 4 LEPTIN TREATMENT INCREASES GROWTH HORMONE SECRETION IN CULTURED GH1 CELLS 77 Introduction............................................................................................................... 77 Methods..................................................................................................................... 79 GH1 Cells.............................................................................................................. 79 Plating Cells and Leptin Concentration................................................................ 79 Five-Day Time-Course ......................................................................................... 79 Media Experiment................................................................................................. 80 RT-PCR for Leptin Receptor ................................................................................ 80 Western Immunoblot for Leptin Receptor ............................................................ 80 GH Radioimmunoassay ........................................................................................ 81 Statistics ................................................................................................................ 81 Results ....................................................................................................................... 81 Ob-Rb mRNA and Protein Expression ................................................................ 81 Five-Day Time-Course ......................................................................................... 82 Media Experiment................................................................................................. 82 Discussion ................................................................................................................. 85 5 LEPTIN RESISTANCE IS ASSOCIATED WITH HYPOTHALAMIC RECEPTOR mRNA AND PROTEIN DOWNREGULATION 88 Introduction............................................................................................................... 88 Methods..................................................................................................................... 89 Animals ................................................................................................................. 89 Groups................................................................................................................... 90 Serum Measurements ............................................................................................ 90 Leptin Challenge Test Pilot Study........................................................................ 91 Leptin Challenge Test........................................................................................... 91 RT-PCR for Leptin Receptor ................................................................................ 91 Western Immunoblot for Leptin Receptor ............................................................ 92 Statistics .................................................................................................................... 92 Results ....................................................................................................................... 93 Leptin .................................................................................................................... 93 Body Weight ......................................................................................................... 93 Food Intake ........................................................................................................... 93 Leptin Challenge Test Pilot Studies...................................................................... 94 Leptin Challenge Test........................................................................................... 94 Leptin Receptor mRNA in Hypothalamus ............................................................ 94 Leptin Receptor Protein Expression in Hypothalamus ......................................... 94 IGF-1 Values......................................................................................................... 95 Hormonal and Metabolic Measures ...................................................................... 95 vii Discussion ............................................................................................................... 100 6 LEPTIN'S EFFECTS ON FOOD INTAKE AND BODY WEIGHT ARE DIFFERENTIALLY ATTENUATED IN RATS FED A LOW-FAT OR HIGHFAT DIET 107 Introduction............................................................................................................. 107 Methods................................................................................................................... 108 Animals and Diets ............................................................................................... 108 Osmotic Pumps, Blood Sampling, and Body Temperature ................................ 109 Leptin and IGF-1 Radioimmunoassays............................................................... 109 Statistics .................................................................................................................. 110 Results ..................................................................................................................... 110 Effects of Diet on Food Intake ............................................................................ 110 Leptin .................................................................................................................. 110 Effects of Diet and Leptin on Food Intake.......................................................... 111 Effects of Diet and Leptin on Body Weight ....................................................... 111 Effects of Diet and Leptin on IGF-1 Values ....................................................... 112 Effects of Diet and Leptin on Organ Weights..................................................... 112 Effects of Diet and Leptin on Body Temperature............................................... 112 Discussion ............................................................................................................... 119 7 GENERAL DISCUSSION 125 REFERENCES 134 BIOGRAPHICAL SKETCH 166 viii LIST OF FIGURES Figure Page Figure 2-1: MgCl 2 (mM) and temperature (°C) optimization..................................................56 Figure 2-2: Cycle optimization...............................................................................................56 Figure 2-3: Southern blot of leptin receptor ...........................................................................57 Figure 2-4: Representative RT-PCR from hypothalamus samples..........................................57 Figure 2-5: Trizol extraction ..................................................................................................58 Figure 2-6: Cell lysis buffer extraction...................................................................................58 Figure 2-7: Protein loading optimization................................................................................61 Figure 2-8: Primary antibody concentration optimization.......................................................61 Figure 3-1: Body Weight ........................................................................................................68 Figure 3-2: Food Intake ..........................................................................................................69 Figure 3-3: Leptin Levels .......................................................................................................69 Figure 3-4: Ob-Rb mRNA......................................................................................................70 Figure 3-5: IGF.......................................................................................................................70 Figure 3-6: GH Profile ...........................................................................................................71 Figure 4-1: Ob-Rb mRNA on GH1 Cells ...............................................................................82 Figure 4-2: Five-Day Time-Course........................................................................................83 Figure 4-3: Media Supplemented with 10% Horse Serum and 2.5% FBS .............................83 Figure 4-4: Serum-Free Media ...............................................................................................84 Figure 4-5: Media Supplemented with 12.5% Charcoal Stripped FBS..................................84 ix Figure 5-1: Leptin Levels Indicate Proper Pump Activity ...................................................... 95 Figure 5-2: Body Weight is Dose-Dependently Decreased by Leptin Treatment.................... 96 Figure 5-3: Food Intake is Initially Decreased by Leptin; Ultimately Resistance Develops... 96 Figure 5-4: Leptin Challenge Test Pilot Study........................................................................ 97 Figure 5-5: Leptin Challenge Test.......................................................................................... 97 Figure 5-6: Leptin Receptor mRNA in Hypothalamus ............................................................ 98 Figure 5-7: Leptin Receptor Protein Expression in Hypothalamus ......................................... 98 Figure 5-8: IGF-1 Values ....................................................................................................... 99 Figure 6-1: Absolute Food Intake Reported in Various Diet Parameters................................113 Figure 6-2: Leptin Levels Indicate Proper Pump Activity ......................................................114 Figure 6-3: The Effects of Leptin on Food Intake in Diets of Varying Calorie and Fat Contents ................................................................................................................115 Figure 6-4: The Effects of Leptin on Food Intake - Normalized to 100 grams Body Weight ..................................................................................................................116 Figure 6-5: The Effects of Leptin on Body Weight in Diets of Varying Calorie and Fat Contents ................................................................................................................117 Figure 6-6: IGF-1 Values .......................................................................................................118 x LIST OF TABLES Table Page Table 1-1: Obesity Models..................................................................................................... 5 Table 1-2: Orexigenic and Anorexigenic Neuropeptides Regulated by Leptin....................... 18 Table 1-3: Leptin Receptors................................................................................................... 23 Table 1-4: Mediators that Inhibit GH Secretion in Both Man and Rodents ............................ 32 Table 1-5: Mediators that Stimulate GH Secretion in Both Man and Rodents........................ 32 Table 3-1: Growth Hormone Profile of Diet-Treated and Control Rats ................................. 71 Table 5-1: Hormonal and Metabolic Measures...................................................................... 99 Table 6-1: Organ Weights ......................................................................................................118 Table 6-2: Body Temperature ................................................................................................119 Table 7-1: Model of GH Regulation by Leptin.......................................................................133 xi Abstract of Dissertation Presented to the Graduate School of the University of Florida in Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy LEPTIN RESISTANCE REDUCES GROWTH HORMONE SECRETION AND CONTRIBUTES TO THE PATHOGENESIS OF OBESITY By Robin Leigh Martin May 2000 Chairman: William J. Millard Major Department: Pharmacodynamics Leptin is secreted by adipocytes in amounts indicative of body fat content. Variations in leptin levels contribute to hypothalamic regulation of feeding and metabolism to maintain body weight homeostasis. Importantly, if there is increasing leptin with increasing body fat and the hypothalamus cannot detect or respond to it, such as is seen in leptin resistance, obesity may develop. Growth hormone (GH) is secreted by the anterior pituitary and directly influences body composition through lipolysis. GH also stimulates the secretion of insulin-like growth factor I (IGF-1), which mediates some of the actions of GH. The hypotheses of this dissertation are (1) under normal circumstances, leptin stimulates GH, and (2) in the leptin resistance situation described above, leptin fails to xii stimulate GH. In obesity, GH is severely attenuated, and leptin resistance may be the mechanism by which this occurs. To test the first hypothesis, rats were given diets with varying fat contents and GH was measured. The diet with the highest fat content did not cause obesity; there were elevated levels of leptin but not enough to produce resistance. These animals had an enhanced GH-IGF-1-axis. In a second study, leptin treatment resulted in elevated GH secretion from a GH-secreting cell line (GH1), further strengthening the hypothesis. To test the second hypothesis, rats were given normal rat chow and implanted with osmotic minipumps for the continuous infusion of two doses of leptin or vehicle. The leptin-treated animals developed resistance, as measured by loss of effect on food intake but not on various metabolic measures, including glucose, triglycerides, and insulin. IGF-1 was attenuated in the high-dose leptin group. Leptin receptors were reduced in this study. A final study utilized both the diets of varying fat content and the leptin-filled osmotic minipumps. Leptin was shown to lose its effectiveness in animals fed the high-fat diet and IGF-1 was attenuated. The rats were fed the diets for a longer period than in the first diet study, allowing time for the development of leptin resistance. IGF-1 was also attenuated in animals infused with leptin. The results of these two studies agree with the second hypothesis. CHAPTER 1 LITERATURE REVIEW Obesity Obesity is one of the most common health problems in industrial societies [Grundy and Barnett 1990]. Obesity is a health threat in part because it is associated with many other diseases, including type II diabetes, gallstones, and certain cancers [Grundy and Barnett 1990] and with excessive mortality [NIH 1985]. Data from National Health and Nutrition Examination Surveys (NHANES) show a high correlation between obesity and risk for coronary artery disease [NIH 1985]. It has also been shown that most obese people experience hypertriglyceridemia and hypercholesterolemia [Grundy and Barnett 1990]. In addition, obesity is a very strong risk factor for hypertension. Obesity is epidemic in the United States (US) [Wilding 1998] and its prevalence is increasing [Grundy and Barnett 1990]. Currently, the criteria for inclusion into the overweight and obese categories are a relationship of height and weight [Bray 1992b]. Body mass index (BMI) is body weight in kilograms divided by height in meters squared. In adults aged 19-34 years, BMI of 1925 kg/m2 is considered normal, 25-30 kg/m2 is overweight, and >30 kg/m2 constitutes obesity. In people 35 years and older, the scale for normal weight is BMI of 19-27 kg/m2, 27-30 kg/m2 is considered overweight, and >30 kg/m2 represents obese. In the US population of adults aged 25 years or more, 42% of men and 28% of women are overweight and 21% of men and 27% of women are obese [Must et al. 1999]. In 1991, 1 2 the prevalence of obesity was 12.0% [Mokdad et al. 1999] and increased to 17.9% in 1998. The incidence of obesity in every state, in both sexes, and in all age groups, races, and educational levels was increased. Nearly 10% of total health care costs are related to obesity [Wilding 1998]. We often blame excessive eating and insufficient exercise for obesity; however there is still no concrete explanation for the fact that some people become obese, despite attempts not to, and others, apparently without much effort, do not [Ravussin and Danforth 1999]. Genetic contributions to the development and maintenance of obesity cannot be eliminated. Studies in twins demonstrated an inherited tendency to gain weight in response to overfeeding [Wilding 1998]. Additionally, it was recently shown that there is unaccounted for physical activity in some people that prevents weight gain [Levine et al. 1999]. This activity has been called NEAT, for nonexercise activity thermogenesis, and is made up of "nonvolitional muscle activity" including maintenance of posture, fidgeting, and muscle tone. In some people, NEAT is triggered by excess food and prevents fat gain. This concept was proposed in the past [Widdowson et al. 1954] with the suggestion that fidgetiness was more important in prevention of weight gain than seemed obvious. It was shown that increased fidgeting enhanced energy expenditure [Rassuvin et al. 1986; Zurlo et al. 1992]. This may account for some of the individual variations observed in food consumption, exercise, and body weight fluctuations. Although obesity is strongly believed to be a risk factor in many diseases, some experts are not in agreement. One professor suggests that obesity is simply a consequence of a lifestyle, such as physical inactivity, that is associated with elevated 3 risks for certain disease, and that obesity in itself is not a risk factor [McDonald 1996]. There are some supporters of this theory, however obesity is intertwined with lack of physical activity and most researchers are in agreement that obesity poses health risks. It has been suggested that, without a change in the progression of obesity in today's society, there will be increasingly overwhelming health care costs and hazardous health conditions related to this disease [Must et al. 1999]. Thus, research relating to obesity is important on an individual as well as on a national level. Obesity Models There are many types of genetic mutations that result in pathophysiological signaling and metabolic alterations and which cause obesity in rodents [Guerre-Millo 1997]. Two such models are the obese (ob/ob) and diabetes (db/db) mice, both of which exhibit hyperphagia, profound early-onset obesity, hyperglycemia, hyperinsulinemia, infertility [Coleman 1973], and defective thermoregulation [Coleman 1978] indicative of a hypothalamic defect. The phenotypes of the ob/ob and db/db mice are identical and the strains can only be distinguished by genetic mapping or through parabiosis studies [Coleman 1973; Coleman 1978]. To study these strains of mice, parabiosis studies were completed in the following pairs of mice: db/db + ob/ob, ob/ob + normal, and db/db + normal. Parabiosis is the surgical joining of two mice in a manner in which they share the same circulatory system. In the db/db + ob/ob parabiosis study, the ob/ob partner became hypoglycemic, lost weight, and starved to death [Coleman 1973]. A similar phenomenon was seen in the normal partner in the parabiosis experiment joining db/db + normal mice [Coleman 1969]. The results of these experiments suggest that there are satiety centers in the ob/ob and normal mice that respond to a circulation factor that is 4 produced by db/db mice. The db/db mice do not respond to the circulating satiety factor, suggesting that the target satiety center is defective [Coleman 1973]. When ob/ob mice were joined with normal mice, there was no alteration in the feeding behavior of the normal mice suggesting that the ob/ob mice do not make the circulating satiety factor in sufficient quantities to affect behavior [Coleman 1973]. The ob/ob genetic defect in mice was first discovered in the laboratory of Snell in 1950 [Ingalls et al. 1950]. Ob/ob is the result of an autosomal recessive mutation [Coleman 1978] of the obese gene located on chromosome 6, which is also the location of the leptin gene [Zhang et al. 1994] (leptin will be discussed in detail later in the chapter). There are two different ob/ob strains of mouse: one with an absence of ob mRNA [Zhang et al. 1994], and the other with 20 times higher expression of a mutant protein [Zhang et al. 1994; Frederich et al. 1995b]. That leptin mRNA is altered in this mutant suggests that the ob gene can be regulated transcriptionally [Mason et al. 1998]. The phenotype of the mice used in this study includes obesity, hyperphagia, low oxygen consumption and body temperature, and reduced physical activity. The db/db genetic defect in mice was first discovered in the laboratory of Coleman in 1966 [Coleman and Hummel 1966; Hummel et al. 1996]. Db/db is the result of an autosomal recessive mutation [Coleman 1978] of the diabetes gene located on chromosome 4, which is the location of the leptin receptor gene [Tartaglia et al. 1995]. The db phenotype results from a mutation of the intracellular portion of the leptin receptor that normally initiates signal transduction [Flier and Elmquist 1997] and as such affects only the long form of the receptor [Guerre-Millo 1997]. Leptin is able to bind 5 specifically and with high affinity to the choroid plexus in db/db mice [Devos et al. 1996], indicating that the short form of the leptin receptor is present. The fa/fa genetic defect in rats was first discovered in the laboratory of Zucker and Zucker in 1961 as an autosomal recessive mutation mapped to chromosome 5 [Truett et al. 1991]. The fa mutation is in the same gene as the db mutation [Chua et al. 1996]. The fa mutation is a missense mutation in the extracellular domain of the leptin receptor [Chua et al. 1996; Flier and Elmquist 1997] that results in inadequate transport to the plasma membrane [Flier and Elmquist 1997]. As in db/db mice, leptin is able to bind specifically and with high affinity to the choroid plexus in fa/fa rats [Devos et al. 1996]. The Koletsky strain of rat develops obesity, hyperinsulinemia, hypertension, and proteinuria [Koletsky et al. 1973] due to a single recessive gene with a mutation in the extracellular domain of the leptin receptor [Takaya et al. 1996]. The same gene as is mutated in the Zucker fa/fa rat is mutated in the Koletsky rat, however, the Koletsky mutation results in a premature stop codon, making this strain of rat a leptin receptor null model. Table 1-1: Obesity Models Model Chromosome Ob/ob mouse 6 Db/db mouse 4 Fa/fa rat 5 Koletsky rat 5 Anomaly Absence of leptin mRNA or expression of mutant protein Mutation of intracellular portion of leptin receptor (long form) Missense mutation of extracellular portion of leptin receptor Premature stop codon – leptin receptor null Reference Ingalls et al. 1950 Coleman and Hummel 1966 Hummel et al. 1966 Zucker and Zucker 1961 Koletsky et al. 1973 6 In addition to genetically occurring models of obesity, obesity models can be created by chemical damage to the hypothalamus. For example, the monosodium glutamate (MSG)-treated animal develops a syndrome in which obesity is its most characteristic feature [Olney 1969]. MSG treatment results in lesions of the brain, specific to those areas surrounding the third ventricle such as the arcuate nucleus of the hypothalamus. Females are infertile and have atrophied ovaries, uteri, and endometria; males can reproduce and have normal testes. MSG rodents gain weight, which is more evident in females, without accompanying hyperphagia, in fact, they appear to be slightly hypophagic [Olney 1969]. The obesity in this model results from metabolic disturbances, which is partially demonstrated in their decrease in body length [Olney 1969]. This decrease in length suggests that there is a defect in growth hormone production, secretion, and/or activity. MSG-treated rats are also lethargic as adults. As a result of damage to the arcuate nucleus, the MSG-treated rodent has elevated leptin mRNA and protein expression compared to control [Frederich et al. 1995b]. Theories of Food Intake Several theories have been proposed to describe the mechanism of initiation of food consumption. Most of the theories revolve around the dual center hypothesis proposed in 1951 by Anand and Brobeck. The dual center hypothesis suggested that the lateral region of the hypothalamus, the "feeding center," played a role in the initiation of feeding and the ventromedial region, the "satiety center," inhibited feeding. This hypothesis was based on studies that showed that lesions to the ventromedial hypothalamus (VMH) and surrounding areas resulted in hyperphagia and obesity [Hetherington et al. 1940] and lesions to the lateral hypothalamus (LH) abolished food 7 intake [Anand and Brobeck 1951]. More recent studies have shown the dual center hypothesis to be too simplistic to be entirely accurate. When lesions of the VMH were restricted solely to that nucleus of the hypothalamus, hyperphagia and obesity did not develop [Reynolds 1963]. In addition, it was shown that hyperphagia developed following injury to other nuclei of the hypothalamus [Jansen and Hutchinson 1969], including areas through which critical catecholamine tracts travel [Sclafani and Berner 1977]. Another study demonstrated that obesity could develop in the absence of hyperphagia [Han 1968]. Although these studies argue against the basic dual center hypothesis, they demonstrate the importance of the hypothalamus in food intake and the development of obesity [Bray and York 1979]. The aminostatic theory of food intake was proposed by Rogers and Leung in 1973. The theory suggests that rats eat less when fed a diet devoid of certain amino acids, a diet with an amino acid imbalance, or a diet high in protein. The authors suggested that brain amino acid content closely resembled the amino acid content in the blood, and that a receptor system, which recognizes blood amino acid concentration, is located in the brain. Their results point to a central involvement in orexic behavior. The glucostatic theory was proposed by Mayer in 1953. The hypothesis was that there are glucose-sensitive receptors, glucoreceptors, in the lateral hypothalamus that initiate feeding when blood glucose is low. Additionally, the hypothesis suggests that there are glucoreceptors in the satiety center of the hypothalamus that terminate feeding when activated by elevated blood glucose. It was discovered that the mere presence of glucose was not sufficient to initiate feeding, but that glucose had to cross the membranes of the glucose-sensitive cells or undergo oxidation. It was later shown that a wide range 8 of metabolites in the blood in addition to glucose are capable of initiating or terminating feeding. The thermostatic theory of food intake was developed by Brobeck in the late 1940s [Brobeck 1948]. The theory states that "animals eat to keep warm and stop eating to prevent hyperthermia." When animals are in warmer temperatures, they will decrease their intake of food to prevent the production of too much heat within the body. Consequently, when animals are in cooler temperatures, food intake increases in an effort to produce heat and prevent hypothermia. It was further hypothesized that the amount of food ingested is dependent on how much heat the food produces and how much heat is required to maintain body temperature. Kennedy argued against this theory [1953] because he felt that the weight loss experienced by these animals was due to the elevated temperatures to which the animals were exposed which may have resulted in dehydration. Kennedy developed his own theory of food intake in 1950 referred to as the lipostatic theory. This theory is similar to Mayer's theory and agrees with the role of the hypothalamus, but utilizes a signal other than glucose. It was theorized that, since young rats are so adept at maintaining constancy of fat stores, there must be some circulating factor that, along with the hypothalamic mechanism, regulates feeding and energy expenditure. The circulating factor was proposed to be some unknown factor of the synthesis, transport, or metabolism of fat. In 1994, a circulating peptide hormone synthesized in fat was discovered [Zhang et al. 1994] which fit the role of Kennedy’s lipostatic factor. This hormone is leptin. 9 Leptin Leptin, coming from the Greek word leptos, meaning thin, is a hydrophobic, 167amino acid, 16-kilodalton protein hormone [Samson et al. 1996] transcribed by the ob gene. Leptin is often referred to as the obese protein. The 4.5 kilobase mRNA from which leptin is translated is 84% homologous between humans and mice and has 67% conservation among a large variety of vertebrates, including human, gorilla, chimpanzee, orangutan, rhesus monkey, dog, cow, pig, rat, and mouse [Zhang et al. 1997]. McGregor et al. [1996] demonstrated that mouse leptin has no homology with other known proteins. Leptin has features of a protein that is secreted [Zhang et al. 1994; Cohen et al. 1996b]. It contains a disulfide bond in the carboxy-terminus [Cohen et al. 1996b]; a mutation of either of the conserved cysteine residues that form the disulfide bond results in the loss of activity [Zhang et al. 1997]. The fragment of leptin that is the predicted signal peptide is 1-21 and fragment 22-56 may be the portion of the protein that is biologically active [Samson et al. 1996]. Additionally, domains between residues 106140 have been shown to induce satiety [Grasso et al. 1997]. Leptin is produced in and secreted mainly from mature white adipocytes. The other types of fat cells (preadipocytes, young adipocytes, fibroblasts, endothelial cells, Schwann cells, and vascular cells) do not express the ob gene [Frederich et al. 1995b; Maffei et al. 1995a]. In culture, leptin mRNA is not expressed until mouse fibroblasts are fully differentiated into adipocytes [Yoshida et al. 1996]. Furthermore, leptin is produced in fat depots throughout the body, including epididymal, parametrial, abdominal, perirenal, and inguinal [Maffei et al. 1995a; McGregor et al. 1996]. Leptin is also expressed in brown adipose tissue [Maffei et al. 1995a], but to a much lesser extent than in white adipose tissue. In addition to expression in adipose tissue, leptin protein or 10 its mRNA have been observed in placenta [Senaris et al. 1997], amniotic fluid [Schubring et al. 1997], and cord blood [Matsuda et al. 1997; Schubring et al. 1997]. Recently, leptin mRNA has also been found in stomach [Bado et al. 1998], skeletal muscle [Wang et al. 1998c], brain [Esler et al. 1998; Wiesner et al. 1999], and pituitary [Jin et al. 1999; Morash et al. 1999; Jin et al. 2000]. Leptin is secreted in proportion to adipose mass [Maffei et al. 1995b; Considine et al. 1996b] and consistent with fat cell size; therefore, heavier humans and animals generally produce more leptin than lean members of the same species do. Leptin is not stored in adipocytes [Rau et al. 1999]. Leptin is secreted in a pulsatile manner with short pulses lasting just over 30 minutes [Licinio et al. 1997]. It has been reported that leptin secretion demonstrates circadian and ultradian rhythms similar to other endocrine hormones [Sinha et al. 1996a; Sinha et al. 1996c]. Leptin's circadian rhythm exhibits increased pulse frequency at night [Sinha et al. 1996a; Licinio et al. 1997] and decreased activity in the afternoon [Sinha et al. 1996a] in both lean and obese humans. Perhaps the purpose of increased leptin at night is to suppress appetite during sleep [Sinha et al. 1996a]. It is interesting to note that this diurnal pattern of leptin secretion is the inverse of that of adrenocorticotropin hormone (ACTH) and cortisol [Licino 1997], however, the apparent diurnal rhythm of leptin seems to be more related to feeding than to time of day [Saladin et al. 1995]. Perhaps leptin helps regulate the diurnal pattern of glucocorticoids [Ahima et al. 1996]. It was suggested that, like other hormones, this pulsatile secretion of leptin might be required for maximum effectiveness [Licino 1997]. Leptin-producing fat depots, however, do not resemble other endocrine glands; they are of varying sizes and are 11 located throughout the body, and as such, would be difficult to coordinate into producing a circadian secretion pattern [Sinha et al. 1996c]. It has been suggested that the apparent pulsatility displayed by leptin actually comes from regulation of its clearance or elimination [Licino 1997]. Upon secretion, leptin distributes throughout the extracellular space and accumulates in cellular water [Cumin et al. 1996] or circulates in the blood either free or complexed with binding proteins [Houseknecht et al. 1996]. Several circulating leptinbinding proteins and a soluble leptin receptor have been observed [Lee et al. 1996]. In lean humans, the majority of circulating leptin is bound [Sinha et al. 1996b]. Perhaps the purpose of leptin binding is to limit bioavailability and regulate its actions on feeding and metabolism homeostasis [Sinha 1997]. Binding proteins also protect leptin from premature degradation or elimination [Liu et al. 1997]. Leptin that is bound remains in circulation and has a half-life of 71 minutes [Hill et al. 1998]. Free leptin is rapidly degraded by proteases and has a half-life of less than 4 minutes [Sharma et al. 1997]. In patients with chronic renal failure [Iida et al. 1996] and end-stage renal disease [Merobet et al. 1997] and in chronic hemodialysis patients [Sharma et al. 1997] leptin levels are elevated, largely represented by the pool of free leptin [Sharma et al. 1997]. In addition, the molecular weight (16-kDa) and half-life (approximately 25 minutes) [Klein et al. 1996] of leptin are similar to other peptide hormones that are degraded by the proximal tubules [Bennett and McMartin 1979], indicating that the kidneys are capable of metabolizing leptin [Klein et al. 1996]. Leptin receptors have been found in the kidneys [Tartaglia et al. 1995] and leptin has, in fact, been shown to have a renal mechanism of elimination [Cumin et al. 1996; Iida et al. 12 1996; Merabet et al. 1997; Sharma et al. 1997]. It was found that leptin is cleared in a two-pool model [Hill et al. 1998] with approximately 75% of leptin being cleared within the first few minutes of secretion [Cumin et al. 1996]. These results, taken together, strongly indicate that leptin is degraded and/or filtered by the kidneys. It is important to note that when pharmacological doses of leptin are administered, the pathways involved in elimination do not become saturated [Cumin et al. 1996; Hill et al. 1998]. Therefore, neither leptin distribution nor half-life differs between lean and fat rats [Vila et al. 1998]. Furthermore, it was shown that malfunctioning clearance or turnover rates [Klein et al. 1996] cannot account for the hyperleptinemic characteristic of obesity. Roles of Leptin Leptin has been implicated as the lipostatic signal that is sensed, directly or indirectly, by the hypothalamus [Campfield et al. 1995; Maffei et al. 1995a; Stephens et al. 1995; Ahima et al. 1996; Levin et al. 1996; Schwartz et al. 1996a; Vaisse et al. 1996; Woods and Stock 1996; Dawson et al. 1997]. Leptin in transported across the bloodbrain barrier by a saturable process [Banks et al. 1996] making it possible that this be the rate-limiting step in leptin bioactivity [Banks et al. 1996; Schwartz et al. 1996b; Golden et al. 1997]. In the hypothalamus, leptin works specifically in nuclei responsible for feeding homeostasis: arcuate, ventromedial, paraventricular, lateral, and ventral premammillary [Mercer et al. 1996]. Leptin is also active in areas of the brain which may be important in controlling metabolism, energy balance, and transport into the brain [Steiner 1996] such as cerebellum, choroid plexus, leptomeninges, cortex, hippocampus, and thalamus [Mercer et al. 1996; Steiner 1996]. 13 Leptin acts to regulate body weight and the size of adipose depots by the regulation of food intake and energy expenditure [Zhang et al. 1994; Campfield et al. 1995; Halaas et al. 1995; Pelleymounter et al. 1995; Halaas et al. 1997]. It has been suggested that leptin is an afferent signal in a negative feedback loop between adipose tissue and the appetite/satiety centers in the brain [Rohner-Jeanrenaud and Jeanrenaud 1996]. Exogenous leptin administration is effective at reducing food intake in animals of normal body weight [Campfield et al. 1995; Halaas et al. 1995] as well as in animals with obesity due to defective leptin production [Pelleymounter et al. 1995]. Daily intraperitoneal leptin injections given to ob/ob mice result in a dose- and time-dependent reduction in food intake and body weight [Pelleymounter et al. 1995]. The effects of leptin on food intake are lost over time, but the effects of leptin on body weight are not. Leptin treatment also restores the oxygen consumption, body temperature, and activity levels in these leptin-deficient mice to levels seen in control wild type mice. When leptin treatment is terminated, body weight levels return to those of control animals [Campfield et al. 1995]. Leptin treatment is also effective when administered continuously via osmotic pumps or when administered centrally [Campfield et al. 1995]. It has been shown that leptin-treated animals lose body weight even when pair-fed the same amount, caloric content, and fat content of food as that consumed by control animals. Factors other than those involved with feeding are implicated in the control of body weight. The rate of metabolism, for example, makes a considerable difference in body composition among individuals, and, as previously mentioned, leptin increases metabolism [Zhang et al. 1994; Campfield et al. 1995; Halaas et al. 1995; Pelleymounter et al. 1995; Halaas et al. 1997]. 14 Brown adipose tissue (BAT) in small mammals is important in heat production and metabolism. When BAT cells are stimulated, mitochondria express uncoupling protein 1 (UCP1) [Cinti 1992]. The sympathetic nervous system activates existing as well as stimulates synthesis of new UCP1 [Zhao et al. 1994]. Upon activation, UCP1 uncouples mitochondria, which results in elevated substrate oxidation [Klingenberg 1990] and thermogenesis. It has been shown that leptin increases energy expenditure and oxygen consumption in rats [Scarpace et al. 1997]; the mechanism by which this occurs is an increase in UCP1 mRNA [Scarpace et al. 1997] via sympathetic activation [Scarpace and Matheny 1998]. In addition, leptin stimulates sympathetic outflow to BAT [Collins et al. 1997]. It has been shown that uncoupling protein 2 (UCP2) can also uncouple mitochondria, and acts in white adipose tissue (WAT) as well as in BAT [Fleury et al. 1997; Scarpace and Matheny 1998]. The mechanism by which leptin stimulates UCP2 does not require sympathetic activation, but rather utilizes some as yet unexplained indirect mechanism [Commins et al. 1999]. Together, these results indicate that leptin activates the sympathetic nervous system and stimulates the production of UCP both directly and by hypothalamic mechanisms, resulting in increased thermogenesis. By reducing feeding and increasing metabolism, leptin decreases fat content. Leptin also reduces fat by other means. Central leptin treatment degrades adipocytespecific genomic DNA in a ladder pattern consistent with apoptosis [Qian et al. 1998]. There is also evidence of condensed chromatin and histological staining consistent with apoptotic events. In addition, leptin inhibits acetyl-CoA carboxylase (ACC) activity [Bai 15 et al. 1996]. ACC is the enzyme that represents the rate-limiting step in fatty acid synthesis. By inhibiting ACC activity, leptin inhibits lipogenesis. Both synthesis and degradation of fat regulate energy stores. In addition to inhibiting lipogenesis, leptin triggers lipolysis by enhancing mitochondrial fatty acid oxidation. This oxidation results in attenuated intracellular fatty acid and triglyceride concentrations [Bai et al. 1996; Qian et al. 1998]. Leptin administration causes weight loss by reduction of fat mass [Halaas et al. 1995] whereas food deprivation alone causes weight loss by reduction of both fat and lean mass. It is clear that leptin is the protein that, consistent with the lipostatic theory of food intake, acts as a homeostatic marker of adipose tissue that regulates the size of adipose mass [Zhang et al. 1994; Frederich et al. 1995b]. Elevated leptin is considered to be a marker of the obese state [Maffei et al. 1995b], but it is also important in the prevention of starvation [Ahima et al. 1996; Spiegelman and Flier 1996; Flier and Elmquist 1997]. Evolutionarily, the role of leptin in starvation may be more important than its role in the prevention of obesity. Leptin is also involved in cardiovascular, renal, and reproductive physiology, and may be part of the circuitry of reward pathways. Regulation of Leptin It has been observed that the downregulation of leptin mRNA expression is dependent on physiologically active leptin receptors [Guerre-Millo 1997]. If the receptor is mutated or otherwise inactive, the result will be overexpression of leptin. Leptin mRNA levels may be regulated by tissue-specific transcription factors or by endocrine and/or paracrine hormonal regulators [Mason et al. 1998]. Regulatory regions of the leptin gene promoter include sites specific for transcription factors that control adipocyte 16 differentiation [Auwerx and Staels 1998]. CCAAT/enhancer-binding protein α (C/EPBα) induces leptin gene expression and peroxisome proliferator-activated receptorγ (PPARγ) inhibits leptin gene expression [Hollenberg et al. 1997]. Leptin levels are acutely altered in response to feeding [Caro et al. 1996b]. Leptin levels are low in food-deprived normal [MacDougald et al. 1995; Hardie et al. 1996; Sinha et al. 1996b] and db/db mice [Frederich et al. 1995b] and rise upon refeeding. Leptin levels can be restored, with leptin treatment, to levels found in fed mice [Ahima et al. 1996]. Human subjects fasted for 24 hours may lose only 0.5% of body fat but 50% of leptin concentration [Boden et al. 1996]. It is clear that leptin expression is regulated in response to nutritional alterations that influence adipose mass [Frederich et al. 1995b]. Leptin and Hormonal Interactions Leptin is intricately intertwined with many central and peripheral hormones. In the brain, leptin affects many neuropeptide systems responsible for the regulation of feeding. Neuropeptide Y (NPY) [Kalra et al. 1989], β-endorphin [McKay et al. 1981], agouti [Lu et al. 1994], agouti-related peptide (AgRP) [Ollmann et al. 1997; Shutter et al. 1997], melanin concentrating hormone (MCH) [Qu et al. 1996], galanin [Sahu 1998], and orexins [Sakurai et al. 1998] stimulate feeding. In general, leptin inhibits peptides that stimulate feeding. Leptin binds to its receptors in the arcuate nucleus and decreases NPY mRNA [Stephens et al. 1995; Schwartz et al. 1996c] thereby attenuating the feeding response normally elicited by NPY. Both agouti [Lu et al. 1994] and AgRP [Ollmann et al. 1997; Shutter et al. 1997] increase feeding by antagonizing a peptide that inhibits feeding, α-melanocyte-stimulating hormone (α-MSH). Agouti and AgRP are decreased 17 by leptin [Mizuno and Mobbs 1999; Wilson et al. 1999]. Galanin [Sahu 1998] and orexins, which act in the lateral hypothalamus to stimulate food intake [Sakurai et al. 1998] are inhibited by leptin [Beck and Richy 1999]. All orexigenic peptides discussed so far are inhibited by leptin. In some cases, however, leptin’s role in the regulation of these peptides is not as clear. Leptin has been shown to increase [Huang et al. 1990] as well as decrease [Sahu 1998] MCH. In addition, leptin receptor mRNA is found on proopiomelanocortin (POMC) mRNAcontaining neurons in the arcuate nucleus [Cheung et al. 1997]. POMC neurons are the precursors of both stimulatory (β-endorphin) and inhibitory (α-MSH) feeding peptides. Leptin has been shown both to decrease POMC mRNA [Schwartz et al. 1997; Thornton et al. 1997] and to increase POMC mRNA [Sahu 1998]. The differential effects of leptin on POMC mRNA may be the result of differential processing of POMC to β-endorphin and α-MSH, respectively. In addition to α-MSH, corticotropin-releasing factor (CRF) [Britton et al. 1982], cocaine and amphetamine-regulating transcript (CART) [Elias et al. 1998; Kristensen et al. 1998], and neurotensin [Sahu 1998] inhibit feeding. Just as leptin tends to inhibit peptides that stimulate feeding, leptin stimulates peptides that inhibit feeding. Leptin increases CRF mRNA in the paraventricular nucleus of the hypothalamus, potentiating the action of CRF on the inhibition of feeding [Schwartz et al. 1996c]. CART, which also is inhibitory on food intake [Elias et al. 1998; Kristensen et al. 1998], is enhanced by leptin [Elias et al. 1998; Kristensen et al. 1998]. Neurotensin is another hypothalamic peptide that inhibits feeding behavior, and leptin increases its gene expression [Sahu 1998]. 18 Table 1-2: Orexigenic and Anorexigenic Neuropeptides Regulated by Leptin Orexigenic Anorexigenic NPY β-endorphin Agouti AgRP CRF MCH Galanin Neurotensin Orexins α-MSH In addition to regulation of central peptides, leptin interacts with peripheral peptides associated with food intake. Blood glucose levels increase directly following a meal and level off between meals. According to the glucostatic theory of food intake [Mayer 1953], glucoreceptors in the lateral hypothalamus initiate feeding when blood glucose is low. Additionally there are glucoreceptors in the satiety center of the hypothalamus that terminate feeding when activated by elevated blood glucose. During food deprivation, glucose levels as well as leptin levels are low. Upon ingestion of a meal, glucose is available to be taken up into cells, and leptin secretion is restored. It has been shown that glucose administration enhances leptin mRNA in mice [Mizuno et al. 1996] and that small glucose infusions following food deprivation prevent the fall of leptin levels in humans [Boden et al. 1996]. Insulin is secreted in response to elevations in blood glucose following a meal. In addition to leptin, insulin would be a potential candidate for the lipostatic theory of food intake [Kennedy 1950; Kennedy 1953] except for one crucial criterion: it is not produced in adipocytes. It is generally well agreed-upon that insulin increases leptin mRNA expression [Yoshida et al. 1996] and leptin production and secretion [Saladin et al. 1995; Mizuno et al. 1996; Wabitisch et al. 1996; Ahren et al. 1997; Barr et al. 1997], but the effects of leptin on insulin are less well defined. There is some controversy about 19 whether leptin increases [Barzilai et al. 1997; Sivitz et al. 1997; Tanizawa et al. 1997], decreases [Cohen et al. 1996a ; Kieffer et al. 1996; Dawson et al. 1997; Poitout et al. 1998], or has no effect [Sinha et al. 1996b] on insulin production, secretion, and/or action. Perhaps these differences in results lie in the fact that leptin and insulin are both dynamically regulated by metabolic factors such as meal ingestion and fasting, and both have an important resistance component, making these comparisons difficult to interpret. Leptin also has multiple hormonal interactions that may or may not be related to orexic behavior. Synthesis and secretion of leptin are increased by glucocorticoids [De Vos et al. 1995; Murakami et al. 1995; Berneis et al. 1996; Slieker et al. 1996; Wabitsch et al. 1996] and leptin has been proposed as an important feedback regulator of the hypothalamic-pituitary-adrenal (HPA) axis [Heiman et al. 1997; Licinio et al. 1997; Pralong et al. 1998]. There is also regulation of leptin by thyroid hormones [EscobarMorreale et al. 1997], growth hormone (GH) [Florkowski et al. 1996], and insulin-like growth factor-1 (IGF-1) [Bianda et al. 1997]. In turn, leptin is required for maximal blood GH levels [Carro et al. 1997]. In fasted mice, estrus is delayed, serum testosterone, luteinizing hormone, and thyroxine levels are reduced, and corticosterone and ACTH are elevated, all of which can be normalized or nearly normalized with leptin treatment [Ahima et al. 1996]. Leptin Receptors Tartaglia et al. [1995] first cloned a high-affinity, single membrane-spanning receptor for leptin mapped to a gene on chromosome 4 with strong sequence homology between mouse and human. The receptor was 894 amino acids in length and the membrane-spanning domain consisted of 23 amino acids. The short intracellular domain 20 of 34 amino acids contained a sequence that allowed binding to Janus protein kinases (JAKs). It was predicted that the receptor was a member of the class I cytokine receptor superfamily and was most closely related to gp130 signaling component of the interleukin-6 (IL-6) receptor, the granulocyte colony-stimulating factor (G-CSF) receptor, and the leukemia inhibitory factor (LIF) receptor [Tartaglia et al. 1995]. Representatives of the class I cytokine family to which the leptin receptor has the highest homology have longer intracellular domains, necessary for signaling, than initially reported by Tartaglia et al. for the leptin receptor. It was later found that the leptin receptor gene creates a splice variant with a longer intracellular domain [Lee et al. 1996] capable of signaling. The gene that encodes the leptin receptor produces multiple splice variants in varying levels in a tissue-specific manner [Lee et al. 1996]. Initially, four membranebound single-gene splice variants of the receptor were found in the mouse: three (Ob-Ra, Ob-Rc, and Ob-Rd) with short intracellular domains originally thought to be incapable of signaling, and one (Ob-Rb) with a longer intracellular domain, through which leptin exerts its biological effects [Lee et al. 1996]. There is also one soluble leptin receptor (Ob-Re). More recently, a new form of the receptor, Ob-Rf, was discovered [Wang et al. 1996]. Ob-Rf has strong sequence homology to the previously cloned isoforms, but with the short intracellular domain dissimilar to the other short forms. Leptin receptors are located in many areas of the body including lung, kidney, liver, ovary, testis, prostate, gastrointestinal tract [Cioffi et al. 1996; Lee et al. 1996], pancreas [Tanizawa et al. 1997] and the central nervous system (CNS). In the CNS, receptors are especially abundant in nuclei of the hypothalamus implicated in feeding homeostasis: arcuate, ventromedial, paraventricular, and ventral premammillary [Mercer 21 et al. 1996]. Leptin receptors have also been observed in cerebellum, choroid plexus, leptomeninges, cortex, hippocampus, thalamus [Mercer et al. 1996; Steiner 1996]. These areas are not necessarily important in modulating feeding behavior, but may be important in controlling metabolism, energy balance, and transport into the brain [Steiner 1996]. Leptin binds with high affinity to the choroid plexus and leptomeninges in rats [Devos et al. 1996]. Ob-Ra is expressed in the choroid plexus (blood-cerebrospinal fluid or blood-CSF barrier), leptomeninges, brain, hypothalamus, testes, adipose tissue [Lee et al. 1996], liver, stomach, kidney, heart, lung [Wang et al. 1996] and in rat brain microvessels that make up the blood-brain barrier [Bjorbaek et al. 1998]. Ob-Ra is thought to help transport leptin across the blood-brain barrier via receptor-mediated transcytosis at the microvessels. It was shown that Ob-Ra on human brain endothelium bind leptin and internalizes it in a temperature-dependent process [Golden et al. 1997]. Leptin may also cross the blood-CSF barrier at the choroid plexus, although this is not the major means by which leptin gains access to the CSF [Bjorbaek et al. 1998]. It has also been suggested that Ob-Ra on the leptomeninges degrades CSF leptin [Bjorbaek et al. 1998] and that Ob-Ra on the choroid plexus clears leptin from the CSF [Tartaglia et al. 1995]. Leptin can be internalized by a coated-pit mechanism by binding to Ob-Ra and partitioned into a lysosomal compartment where it can then be degraded [Uotani et al. 1999]. In addition, Ob-Ra is the receptor in the kidney thought to play a role in clearance of leptin from the circulation [Cumin et al. 1996], possibly by glomerular filtration. Ob-Rb is highly expressed in the hypothalamus [Lee et al. 1996] and is the isoform of the receptor through which leptin exerts a biological effect [Tartaglia et al. 1995]. The hypothalamus is the only tissue in which the long form of the leptin receptor 22 is more abundantly expressed than the short form [Tartaglia 1997]. Within the hypothalamus, Ob-Rb is strongly immunoreactive in the arcuate, paraventricular, surpraoptic [Matsuda et al. 1999], ventromedial, and dorsomedial nuclei [Mercer et al. 1996]. Ob-Rb is also found in brain, cerebellum, testes, adipose tissue [Lee et al. 1996], and pituitary [Jin et al. 1999; Jin et al. 2000]. In pituitary, leptin receptor is found in 70% of ACTH cells, 21% of GH cells, 33% of FSH cells, 29% of LH cells, 32% of TSH cells, and 3% of prolactin cells [Jin et al. 1999]. Recently, a soluble isoform of the leptin receptor (Ob-Re) has been characterized which has an affinity similar to that of the membrane-bound Ob-Rb [Liu et al. 1997]. It has been proposed that the role of Ob-Re is to regulate leptin bioactivity by protection from degradation, slow the clearance process, and inhibit binding to Ob-Rb [Liu et al. 1997]. This soluble receptor is potentially the same protein as a plasma binding molecule [Liu et al. 1997] and has been shown to bind the majority of circulating leptin in mice and humans [Houseknecht et al. 1996]. The other short forms of the leptin receptor are far less characterized. Ob-Rc and Ob-Rd are expressed in heart, testes, adipose tissue, and spleen [Lee et al. 1996]. Ob-Rf is localized in the brain, liver, stomach, kidney, lung, heart, thymus, spleen, and hypothalamus [Wang et al. 1996]. More work needs to be done to elucidate the roles these receptors play in leptin bioactivity. 23 Table 1-3: Leptin Receptors Receptor Length Localization Ob-Ra Short Choroid plexus, leptomeninges, brain microvessels, brain, hypothalamus, testes, adipose tissue, liver, stomach, kidney, heart, lung Ob-Rb Long Ob-Rc and Ob-Rd Ob-Re Ob-Rf Short Short Short Hypothalamus, brain, cerebellum, pituitary, testes, adipose tissue Heart, testes, adipose tissue, spleen Soluble - circulates in blood Brain, hypothalamus, liver, stomach, heart, kidney, lung, thymus, spleen Roles Transports leptin across blood brain barrier, degrades and/or clears leptin from cerebrospinal fluid, clears leptin from circulation Biological effects ? Regulates leptin bioactivity ? Leptin Signaling Members of the class I cytokine receptor superfamily, to which the leptin receptor belongs, individually demonstrate no signaling capacity, but rather require complexation with protein kinases to initiate a signal transduction cascade [Tartaglia et al. 1995]. All membrane-bound isoforms of the leptin receptor have a Box 1 domain [Murakami et al. 1991] and the long form also has a Box 2 domain [Lee et al. 1996; Bjorbaek et al. 1997]. Both Boxes 1 and 2 are required for JAK (Janus kinase or just another kinase) binding and activation of signal transduction via the signal transducers and activators of transcription (STAT) pathway [Murakami et al. 1991]. It was initially suggested that the short forms of the leptin receptor were incapable of signal transduction [Tartaglia et al. 1995]. As stated previously, cytokine receptors cannot initiate a signal transduction cascade independently [Tartaglia et al. 1995]. Many cytokine receptors are capable of 24 signaling following receptor homooligomerization, and it was predicted that Ob-Rb followed the same pattern [White et al. 1997]. It was shown that the long form as well as the short form of the leptin receptor homodimerizes spontaneously [White and Tartaglia 1999] and can form heterodimers upon ligand binding. The JAK-STAT pathway is common in cytokine signaling [Darnell 1997; Pellegrini and Dusanter-Fourt 1997]. JAKs are constitutively associated with the cytoplasmic domain of cytokine receptors. When the appropriate ligand binds to its receptor, the receptor dimerizes and JAKs are able to phosphorylate each other. Upon this activation, JAKs phosphorylate the receptor and various intracellular transcription factors. One such factor is STAT. Src homology 2 (SH2) domains, found on all STATs, promote binding of STATs to phosphorylated receptors [Darnell et al. 1994; Ihle and Kerr 1995; Schindler and Darnell 1995], which subsequently results in phosphorylation of STATs by JAKs. Phosphorylated STATs dimerize, translocate to the nucleus, and induce gene transcription to produce a biological effect. The long form of the leptin receptor has 3-5 sites where tyrosine phosphorylation may occur [Bjorbaek et al. 1997]. JAKs phosphorylate these sites and allow the interaction of STATs with the receptor. Leptin is also capable of signal transduction through pathways that involve the mitogen activating protein (MAP) kinase and insulin receptor substrate I (IRS-1) [Bjorbaek et al. 1997; Murakami et al. 1997; Yamashita et al. 1998]. JAK activation is required to initiate the MAP kinase transduction cascade [Wang et al. 1995; Winston and Hunter 1996]. JAK phosphorylates Shc, which then activates Ras, and the cascade is initiated. Alternatively, JAK mediates IRS-1 phosphorylation [Argetsinger et al. 1995; Johnston et al. 1995], thereby activating Ras. Recently, however, it was found that the 25 short form of the leptin receptor, using the Box 1 motif, was capable of signal transduction through a pathway in which Box 2 was not involved [Bjorbaek et al. 1997; Murakami et al. 1997; Yamashita et al. 1998]. The Box 1 pathway phosphorylates JAK2 and IRS-1 tyrosine residues and activates the MAP kinase cascade, but is incapable of STAT activation. The long form of the leptin receptor was better able to activate the STAT pathway than the short form, however, it was clear that the short form had signaling capabilities [Bjorbaek et al. 1997]. JAK-STAT transcription activity is rapid and transient [Shuai et al. 1992], indicating that the pathway is negatively regulated. Suppressors of cytokine signaling (SOCS) are proteins that are encoded by genes that are activated by the STATs [Yoshimura et al. 1995; Starr et al. 1997]. There are many members of the SOCS family, including SOCS-1 through SOCS-7 and CIS (cytokine inducible SH2-containing protein) [Hilton et al. 1998]. There is differential expression of SOCS in various tissues [TolletEgnell et al.1999] and each member acts in a different way to negatively feedback on JAK or STAT to limit the intensity and/or duration of activation [Nicholson and Hilton 1998; Starr and Hilton 1998]. Leptin treatment has been shown to induce SOCS-3 and CIS mRNA in many target tissues [Emilsson et al. 1999]. Excess leptin production, such as that in obesity, may result in overproduction of cytokine suppression proteins, and as such is a potential mechanism of leptin resistance [Emilsson et al. 1999]. Leptin Resistance As a circulating factor informing the brain of the body’s energy stores, leptin plays a role in the maintenance of body weight homeostasis. Mice with mutations of the ob gene have reduced levels of circulating leptin and exhibit hyperphagia and obesity. 26 When these animals are given exogenous leptin, their body weight is significantly reduced. Mice with mutations of the db gene have elevated levels of circulating leptin but, like the ob mutants, experience hyperphagia and obesity. Leptin treatment in the db mutants is ineffective. Human obesity is not often the result of a mutation, as far as is currently known, so comparisons with these animal models is not entirely accurate; however most obese humans exhibit dramatically elevated levels of circulating leptin [Zhang et al. 1994; Maffei et al. 1995b] and ob mRNA [Considine et al. 1996b]. In this respect, humans more closely resemble the db/db mouse than the ob/ob mouse and, in general, are insensitive to the effects of leptin [Frederich et al. 1995a; Maffei et al. 1995b; Halaas et al. 1997]. The problem in humans is not the production of leptin, but the response to leptin. When the brain is unable to respond to elevated leptin levels, it is referred to as resistance [Flier and Elmquist 1997]. It has been suggested that this so-called leptin resistance is a major cause of obesity [Frederich et al. 1995a; Frederich et al. 1995b; Maffei et al. 1995b; Considine et al. 1996b]. There are many potential mechanisms of leptin resistance. Transport of leptin across the blood-brain barrier is unidirectional from blood to brain [Banks et al. 1996] and occurs by a saturable process [Caro et al. 1996a]. The saturable transport of leptin into the brain may be a critical component in resistance [Tartaglia 1997]. In a recent study in mice, it was shown that central administration of leptin to peripherally resistant animals resulted in significant reductions in food intake and body weight [Van Heek et al. 1997], suggesting that leptin may lose its ability to cross the blood-brain barrier in the resistant state. In addition, it was shown that the cerebrospinal fluid (CSF)-to-serum leptin ratio is lower in obese humans compared to 27 lean [Caro et al. 1996a; Schwartz et al. 1996b]. Other investigators have also demonstrated an inability of leptin to cross the blood-brain barrier in resistant animals [Banks et al. 1996; Caro et al. 1996a; Schwartz et al. 1996b; Van Heek et al. 1997]. However, all aspects of leptin resistance cannot be explained by its inability to cross the blood-brain barrier. The Koletsky rat, a leptin receptor null model, has leptin CSF levels similar to controls [Wu-Peng et al. 1997], indicating that leptin can enter the brain independently of leptin receptors and other transport mechanisms [Bjorbaek et al. 1998]. Another mechanism of leptin resistance may be explained by the inability of the hypothalamus to detect leptin [Tartaglia 1997] due to defective hypothalamic leptin receptors [Considine et al. 1996a ; Dawson et al. 1997]. Considine et al. report that the full-length leptin receptor is expressed in the human hypothalamus [Considine et al. 1996a]; perhaps leptin receptors are functional but downregulated in response to hyperleptinemia. The possibility that there is a blocked leptin receptor has also been suggested [McGregor et al. 1996]. Alternatively, leptin resistance may occur downstream of the receptor in the signal transduction pathway [Tartaglia 1997]. There are varying stages of leptin resistance among different strains of obese mice [Halaas et al. 1997]. In a study comparing many strains of obese mice, leptin given peripherally was effective in lean mice and in one strain of obese mice but at a much higher dose. In addition, leptin given directly into the brain was effective in two of three strains of obese mice tested but at dramatically different doses. The significance of this study is that there are likely to be variations of leptin resistance in obese humans as well. 28 Human Leptin Mutations Although the majority of humans experience obesity due to leptin resistance [Frederich et al. 1995a; Frederich et al. 1995b; Maffei et al. 1995b; Considine et al. 1996b], there are some instances of mutations of the leptin or leptin receptor. Two cousins in a Pakistani family, normal weight at birth, developed severe early-onset obesity [Montague et al. 1997]. No one in the families of either of the children was obese. It was found that the obesity is due to a deletion mutation of a single nucleotide of the leptin region of the obese gene, which results in a premature stop codon. Both children are homozygous for the mutation; their parents are heterozygous. This mutation caused the creation of a form of leptin that cannot be secreted; serum leptin levels are nearly undetectable. In addition, the low levels of serum leptin present lack the disulfide bond that produces bioactivity [Rau et al. 1999]. The children are hyperphagic but display no noticeable impairment of basal energy expenditure [Montague et al. 1997]. Mutations of the obese gene, as have been described here, are rare [Reed et al. 1996]. It has been shown, however, that mutations on chromosome 6 in the region of the obese gene predispose toward extreme obesity [Clement et al. 1996]. In a Turkish family, congenital leptin deficiency resulted in severely obese individuals. This deficiency is the result of a missense mutation in the leptin gene [Strobel et al. 1998], which results in impaired secretion of leptin. The family members homozygous for the mutation are severely obese, hyperphagic, and have low sympathetic tone. The mutations in both the Pakistani and Turkish families are identical to that in the ob/ob mouse. A Kabilian family with severely obese individuals are homozygous for a nonsense mutation in the leptin receptor [Clement et al. 1998]; this mutation being homologous to 29 that in the db/db mouse and fa/fa rat. The mutation results in a protein that lacks the transmembrane and intracellular domains of leptin receptor. These subjects develop obesity within the first year of life, are hyperphagic, and have high circulating levels of leptin. As in the Pakistani and Turkish families, the members of the Kabilian family who are heterozygous for the mutation do not develop the phenotype. Leptin Treatment in Humans and Leptin Gene Therapy As would be predicted from studies in which leptin was administered to ob/ob mice, leptin treatment has been shown to be effective in an individual with congenital leptin deficiency. The nine-year old female cousin from the Pakistani family was treated with recombinant leptin for 12 months [Farooqi et al. 1999]. The treatment resulted in weight reduction due to loss of fat and negative energy balance due to reduced food intake. After 2 months of therapy, leptin antibodies were detectable in the plasma; they did not, however, appear to interfere with the response to the treatment. Leptin treatment may also be effective in patients whose obesity is not the result of genetics. In a clinical trial in which leptin was given to both obese and lean subjects [Heymsfield et al. 1999], leptin was effective at producing weight loss in a dosedependent manner over a 4-week period. In addition, leptin was effective for 24 weeks in obese subjects. Weight loss was primarily due to fat loss [Heymsfield et al. 1999], as was seen in the Pakistani female [Farooqi et al. 1999]. The results of this study are promising because they indicate that, in instances in which resistance is not the result of a mutation, high doses of exogenously administered leptin may overcome leptin resistance and help obese individuals lose weight [Heymsfield et al. 1999]. 30 It has been shown that leptin gene therapy is effective in animals [Chen et al. 1996; Murphy et al. 1997; Shimabukuro et al. 1997]. In animals that were made hyperleptinemic with an adeno-associated virus (AAV), triglyceride content in several tissues which express leptin receptor was reduced [Shimabukuro et al. 1997]. Murphy et al. [1997] created a recombinant AAV, which expresses leptin both in vivo and in vitro. A single intramuscular injection of this rAAV to ob/ob mice resulted in correction of metabolic defects, obesity, diabetes, elevated food intake and body weight, hyperinsulinemia, and insulin resistance for as long as six months [Murphy et al. 1997]. Chen et al. [1996] also induced chronic hyperleptinemia in normal adult male rats by the aid of adenovirus-mediated gene delivery. In these animals, there was a reduction in food intake, no increase in body weight, and disappearance of adipose tissue. These results, taken together with results from leptin treatment in humans, suggest that leptin gene therapy may be beneficial in the treatment of human obesity. Growth Hormone Growth hormone (GH) is a 21.5 kilodalton, 191 amino-acid polypeptide that is secreted episodically from the somatotropic cells of the anterior pituitary gland [Martin et al. 1978]. The secretion of GH is under direct hypothalamic control mediated by two neuropeptides: growth hormone releasing hormone (GHRH) and somatotropin-release inhibiting hormone (SRIH) [Martin and Millard 1986; Millard 1989]. GHRH, synthesized and released from neurons located in the hypothalamic arcuate nucleus, is responsible for the high amplitude GH secretory episodes in both man and rats. SRIH, on the other hand, mediates the prolonged intervals whereby very low levels of GH secretion occur. SRIH neurons are confined to the periventricular nucleus of the hypothalamus 31 [Kiyama and Emson 1990] in both man and rodents. GHRH and SRIH are secreted consistently into hypophyseal portal blood, and each also display a periodic surge every 3-4 hours. Each neuropeptide is secreted 180° out of phase with the other [Tannenbaum and Ling 1984]. SRIH regulates GHRH secretion [Tannenbaum 1994] and the release of GHRH and SRIH are, in turn, regulated by a host of other neuropeptides and putative neurotransmitters, including neuropeptide Y (NPY). In addition, synthetic growth hormone releasing peptides (GHRPs) have been shown to be more potent at stimulating GH release than GHRH [Bowers et al. 1991; Bowers 1994]. GHRPs synergize with GHRH to release GH, which suggests that GHRH and GHRPs act at different pituitary somatotrope receptors and through different second messenger pathways. GHRPs also regulate GH by antagonizing the actions of SRIH [Ghigo et al. 1997]. In addition to regulation by GHRH, SRIH, and GHRP, GH secretion is altered in response to many other neuropeptides, neurotransmitters, hormones, and physiological conditions (Tables 1-4 and 1-5). These influences may occur directly at the level of the pituitary or by control of the hypothalamic regulators of GH. When considered to its full extent, the regulation of GH secretion is very complex. To further complicate the matter, the regulation of GH is not always similar among species or within various disease states. For example, the regulation of GH by histamine [Netti et al. 1981; Knigge et al. 1990], bombesin [Pontiroli and Scarpignato 1986; Scarpignato et al. 1986; Benitez et al. 1990], neuromedin C [Houben and Denef 1991], excitatory amino acids [Mason et al. 1983], starvation [Shibasaki et al. 1985], and exercise [Felsing et al. 1992; Butkus et al. 1995] have been demonstrated only in rats or have differential effects in man and rodents. In addition, regulation of GH by thyrotropin releasing hormone [Czernichow et al. 1976; 32 Mueller et al. 1977; Valentini et al. 1989; Giustina et al. 1995] and dopamine [Liuzzi et al. 1974; Chihara et al. 1979; Peillon et al. 1979; Kitajima et al. 1986; Schober et al. 1989] is altered in different disease states. Table 1-4: Mediators that Inhibit GH Secretion in Both Man and Rodents Mediator Type Reference SRIH Neurohormone Arginine vasopressin Neurohormone Martin and Millard 1986; Millard 1989 Martin et al. 1978a β2 adrenergic agonists Neurotransmitter Mauras et al. 1987 Nicotinic muscarinic agonists Plasma fatty acid concentration Age Neurotransmitter Mendelson et al. 1981 Physiological condition Physiological condition Physiological condition Imaki et al. 1985 Obesity Iranmanesh et al. 1994; Veldhuis and Iranmanesh 1996 Iranmanesh et al. 1994; Veldhuis and Iranmanesh 1996 Table 1-5: Mediators that Stimulate GH Secretion in Both Man and Rodents Mediator Type Reference GHRH Neurohormone Martin and Millard 1986; Millard 1989 GHRP Synthetic Bowers et al. 1980; oligonucleotides Bowers et al. 1984 Galanin Neuropeptide Murakami et al. 1989; Giustina et al. 1992 Opiates Neuropeptides Delitala et al. 1983; Murakami et al. 1985 Neurotransmitter Lancranjan and Marback 1977; α2 adrenergic agonists Miki et al. 1984 Cholinergic muscarinic Neurotransmitter Locatelli et al. 1986 agonists Serotonin Neurotransmitter Imura et al. 1973; Murakami et al. 1986 GABA Neurotransmitter Cavagnini et al. 1977; Acs et al. 1987 Testosterone Hormone Veldhuis et al. 1995a; Mauras et al. 1996 33 GH pulses can be detected in the blood every 3-5 hours in humans, baboons [Steiner et al. 1978], monkeys [Quabbe et al. 1981], rabbits, dogs, and rats [Martin 1978]. There are binding proteins for GH found in the plasma. These proteins, GHBPs, correspond to the extracellular domain of the GH receptor [Leung et al. 1987] and, as such, bind GH with high affinity. The role of these circulating binding proteins is to protect GH from premature degradation or clearance [Baumann et al. 1987] and thus increase its half-life, which is 17-45 minutes [Martin et al. 1978]. GH secretion is associated with the onset of sleep and slow wave sleep [Goldstein et al. 1983] and levels fluctuate throughout the life span. GH levels are very high in neonates due to elevated daytime and nighttime bursts of GH [de Zegher et al. 1993]. Values fall shortly after birth and remain stable from day to day in prepubertal children [Martha et al. 1996]. During puberty, pulsatile GH secretion is amplified as much as 3fold [Mauras et al. 1996]. Following puberty, young adults experience a fall in GH to levels equal to or lower than those in the prepubertal stage [Martha et al. 1996]. These levels slowly fall throughout adulthood and are greatly reduced in aged individuals [Veldhuis and Iranmanesh 1996]. GH has many important roles in the body. It is mitogenic, increasing RNA and DNA synthesis and inducing cell division [Merimee 1979]. It is anabolic, incorporating amino acids into proteins, increasing muscle size and strength, and lengthening and widening bones [Merimee 1979]. GH also regulates body composition through nitrogen sparing and lipolysis [Ho et al. 1996] directly at the level of the adipocyte [Fagin et al. 1980; Vikman et al. 1991]. In obese animals, GH burst amplitudes are reduced [Veldhuis et al. 1991] and the resulting (decreased) levels of GH may not be sufficient to affect 34 lipolysis. Ahmad et al. found that in genetically obese Zucker (fa/fa) rats, GH and GHRH and their messages were decreased by the age of five weeks [Ahmad et al. 1993]. When the rats were given recombinant human GH (rhGH), a decrease in body weight was observed. There are various signaling pathways by which GH works, but the main pathways are JAK-STAT [Argetsinger and Carter-Su 1996] and SOCS [Tollet-Egnell et al.1999]. Once secreted into the plasma, GH stimulates the release of insulin-like growth factor I (IGF-1). IGF-1 is released in a continuous manner and circulates in the plasma bound predominantly to one of its binding proteins, IGF binding protein-3 (IGFBP3) [Lamson et al. 1991]. Both IGF-1 and its binding protein have relatively long half-lives in this complex [Mohan and Baylink 1996]. Since GH stimulates IGF-1 and because of its continuous release and relatively long half-life, IGF-1 is considered a good indicator of GH status over time [Blum et al. 1990]. IGF-1 does not appear to have circadian rhythmicity, so time of sampling is not important [Sara and Hall 1990]. The main source of circulating IGF-1 is the liver [Sara and Hall 1990]. Regulation of liver IGF-1 is strongly regulated by GH, but fasting and refeeding also have IGF-1 regulatory roles [Philipps et al. 1989]. IGF-1 is also produced in many other tissues of the body in which it acts as a paracrine factor [Hall and Bozovic 1969]. GH is a primary regulator of IGF-1 in extrahepatic tissues such as heart, lung, and pancreas [Roberts et al. 1987]. Other IGF1 regulators in extrahepatic tissues include prolactin [Murphy et al. 1988] and various growth factors and trophic hormones [Clemmons and Shaw 1983; Clemmons 1985; Sara and Hall 1990]. IGF-1 is also produced locally in response to neural [Hansson et al. 1986], arterial [Hansson et al. 1987], and skeletal muscle injury [Jennische et al. 1987]. 35 GH has both direct effects in the body and indirect effects mediated by IGF-1. Some of the direct effects of GH include lipolysis [Fain et al. 1965] and gluconeogenesis [Dawson and Hales 1969]. Some of the effects mediated by IGF-1 include many of the growth-promoting effects in muscle, cartilage, and bone, such as DNA synthesis, cell proliferation, and protein synthesis [Schoenle et al. 1982]. Green et al. [1985] proposed a dual model of GH action which suggested that GH stimulates differentiation of precursor cells and IGF-1 then stimulates growth. Excess of Deficiency of Growth Hormone Excess and deficient GH secretion result in pathological conditions that can be corrected pharmacologically. For example, GH hypersecretion usually occurs as the result of a pituitary adenoma [Hansen et al. 1994]. When this hypersecretion begins before puberty, gigantism occurs. When the hypersecretion starts after puberty, the result is acromegaly. In both conditions, individuals experience elevated IGF-1, insulin resistance, and impaired glucose tolerance [Quabbe and Plockinger 1996]. There is also increased fat mobilization [Weil 1965], amino acid retention, and stimulated protein synthesis [Russell-Jones et al. 1993], which result in decreased fat mass and increased lean mass [Salomon et al. 1993]. Muscle strength is not necessarily greater. Increased total body water and extracellular fluid result in mild arterial hypertension [Quabbe and Plockinger 1996]. Bone turnover is increased [Lieberman et al. 1992]. In gigantism, bones are widened as well as lengthened. In acromegaly, the GH excess occurs after puberty when the epiphyseal plates fuse and bones can no longer grow in length. A thickening of bones occurs in acromegaly. In addition, there is excess growth of cartilage and soft tissue cell mass [Quabbe and Plockinger 1996]. Hypersecretion of GH can be 36 treated long-term with an analogue of SRIH, octreotide [Sassolas 1992]. Octreotide therapy reduces GH and IGF in many patients with acromegaly [Hansen et al. 1994] and these reductions result in normalization of lean body mass, body fat, extracellular water content [Bengtsson et al. 1989; Bengtsson et al. 1990], and joint pain [Sassolas 1992]. However, body cell mass remains high [Bengtsson et al. 1990] and there is an increased incidence of gallstones [Sassolas 1992]. Individuals deficient in GH experience increased truncal fat mass and decreased lean mass [DeBoer et al. 1992]. There are also symptoms of osteopenia [Holmes et al. 1994], adverse lipid profiles [Cuneo et al. 1993], glucose intolerance and resistance [Beshyan et al. 1994; Johansson et al. 1995], and reduced exercise capacity [Nass et al. 1995]. Finally, there is a general reduction in the quality of life [Bjork et al. 1989; McGauley 1989; Rosen et al. 1994]. Growth hormone replacement increases circulating IGF-1 values. It normalizes lean and fat mass [Binnerts et al. 1992], redistributing adipose tissue from abdominal to peripheral depots [Bengtsson et al. 1992]. Patients gain muscle [Bengtsson et al. 1990] through increased protein synthesis. There are also increases in bone turnover [Binnerts et al. 1992]. In addition, individuals undergoing GH replacement therapy experience improved quality of life [McGauley et al. 1990] and cognitive functioning [Almqvist et al. 1986]. Side effects can include transient water retention [Binnerts et al. 1992] that can be eliminated by lowering the dose of GH. Growth Hormone and Obesity It is widely recognized that GH is attenuated in obesity. A number of negative correlations have been demonstrated between measures of body mass and GH, including BMI vs. GH release, amplitude, and half-life [Iranmanesh et al. 1991; Veldhuis et al. 37 1995b] and percent body fat vs. GH release and half-life [Veldhuis et al. 1995b]. The somatotrope secretory capacity is reduced in obesity [Maccario et al. 1997]; obese individuals experience reductions in frequency, amount, and duration of GH secretion and shorter plasma GH half-life [Veldhuis et al. 1991]. The exact mechanism(s) of the reduction of GH in obesity is unknown [Scacchi et al. 1999], however, it is known that there are multiple players involved. There may be a hypothalamic contribution to attenuation of GH in obesity. It was previously suggested that elevated SRIH contributed to attenuated GH in obesity [Cordido et al. 1989; Tannenbaum et al. 1990]. However, another study showed a decreased CSF level of SRIH in obese patients [Brunani et al. 1995]. It was also previously hypothesized that reductions in GHRH caused the GH attenuation in obesity [Tannenbaum et al. 1990; Ahmad et al. 1993]. This hypothesis was also challenged when it was demonstrated that GHRH concentrations did not differ between obese and nonobese subjects [Brunani et al. 1995]. However, when given exogenous GHRH, overweight subjects exhibit a blunted rise in GH whether GHRH was given by intravenous bolus [Williams et al. 1984], continuous intravenous infusion [Davies et al. 1985], or pulsatile intravenous administration [Kopelman and Noonan 1986]. In addition, the response of GH to GHRPs was reduced in obese patients compared to lean [Cordido et al. 1993]. Obviously, more work needs to be done to fully elucidate the role of the hypothalamic regulatory peptides on GH in the obese state. Another potential mechanism of attenuated GH in obese patients is decreased half-life, which may be explained by the reduction of circulating GHBPs in obese patients [Hochberg et al. 1992; Argente et al. 1997; Kratzsch et al. 1997]. There is a 38 positive correlation between both BMI [Hochberg et al. 1992] and percent body fat [Kratzsch et al. 1997] and GHBPs in obesity. GHBPs act to protect GH from degradation and thus extend its half-life, so it is logical that reductions in the amount of circulating GHBPs result in shorter GH half-life. In addition, metabolic clearance rate of GH is increased in obese individuals [Veldhuis and Iranmanesh 1996]. Obese individuals also demonstrate a reduced number of IGF-1 receptors and reduced binding [Hochberg et al. 1992]. There is some controversy as to the effects of obesity on IGF-1 levels. There have been studies demonstrating both reduced [Argente et al. 1997] and elevated [Hochberg et al. 1992] plasma IGF-1 levels in obesity, but reductions in receptor binding limit the activity of IGF-1 either way. There are many circulating factors that feedback on GH to reduce its secretion and which may play a role on the effects of GH in obesity. For example, it is known that circulating insulin feedsback negatively on GH secretion [Lanzi et al. 1997]. Since obesity is associated with hyperinsulinemia [Polonsky et al. 1988], it is tempting to speculate that this negative feedback may play a role in inhibiting GH. However, there are diseases in which insulin is elevated and GH is normal [Maccario et al. 1996]. In addition, reduction of hyperinsulinemia in obesity does not normalize GH [Chalew et al. 1992]. Non-esterified fatty acids (NEFA) are another example of circulating factors that feedback to inhibit GH. Upon lipolysis, NEFA are released into circulation. This increase in NEFA has been shown to negatively feedback at both the level of the pituitary [Casanueva et al. 1987] and the level of the hypothalamus to reduce GH secretion. Obese individuals exhibit elevated plasma NEFA [Opie and Walfish 1963; Golay et al. 1986], adding another potential mechanism for the attenuation of GH in obesity. 39 Promisingly, the attenuation of GH is obesity is lessened with the reduction in body weight. Weight loss restores both spontaneous and GHRH-stimulated GH release [Williams et al. 1984] and normalizes elevated GHBP levels [Rasmussen et al. 1996; Argente et al. 1997]. In addition, GH treatment of obese subjects results in desirable effects on body mass [Jorgensen et al. 1994; Richelsen et al. 1994]. Leptin and Growth Hormone It has recently been shown that circulating leptin and GH levels are related. Studies demonstrated a normalization of elevated leptin levels upon GH replacement in GH-deficient adults [Florkowski et al. 1996, Fisker et al. 1997], most likely a consequence of the decrease in body fat which occurred as a result of the lipolytic effects of GH. In addition to regulation of leptin by GH, there is regulation of GH by leptin. Leptin induces spontaneous and GHRH-induced GH secretion [Tannenbaum et al. 1998], at least partially by inhibiting SRIH release. Carro et al. [1997] demonstrated that leptin antiserum decreased GH amplitude and nadir in rats suggesting that normal levels of leptin are required for normal GH secretion. They also showed that in fasted animals with low leptin and low GH, administration of exogenous leptin normalized GH secretion. The effect of leptin on GH may occur at the hypothalamic level. Leptin receptors colocalize with GHRH in neurons [Hakansson et al. 1998] and are also found in the periventricular nucleus [Mercer et al. 1996] where SRIH neurons are located [Kiyama and Emson 1990]. When administered to fasted rats, leptin prevented the inhibition of GHRH mRNA that is normally seen in conjunction with fasting [Dieguez 1998; Carro et al. 1999]. When incubated with rat hypothalamic neurons, leptin decreased basal SRIH 40 mRNA and secretion [Quintela et al. 1997]. The regulation of either, or both, of these two hypothalamic GH regulatory factors by leptin may result in indirect regulation of GH by leptin. A direct regulation of GH by leptin may also occur at the pituitary level. It has recently been shown that leptin and leptin receptors are produced in the anterior pituitary of humans [Jin et al. 1999] and rodents [Jin et al. 2000]. One potential mechanism by which leptin may regulate GH is via neuropeptide Y (NPY). NPY is a 36-amino acid peptide isolated in 1982 by Tatemoto et al. that has considerable evolutionary conservation [Tatemoto et al. 1982]. NPY is the neuropeptide most abundantly found in the brain [Chronwall et al. 1985] with high densities in and complex networks throughout the hypothalamic nuclei, including the arcuate, paraventricular, and periventricular nuclei [Chronwall et al. 1985; DeQuait and Emson 1986]. NPY neurons often colocalize with hormones and norepinephrine. In the arcuate nucleus, NPY neurons colocalize with GHRH and in many other brain areas, NPY and SRIH neurons colocalize [Vincent et al. 1982; De Quiat and Emson 1986; Fuxe et al. 1989; Okada et al. 1993]. NPY plays roles in reproduction [Kalra et al. 1989], circadian rhythmicity, and cardiovascular control [Tatemoto 1989], but perhaps the stimulation of feeding is the best know function of NPY. When given centrally, NPY invokes a robust food intake in male and female rats [Kalra et al. 1989] during normal nighttime feeding and during daylight [Clark et al. 1985]. The response is observed within 15 minutes, and is dose-dependent. NPY specifically increases the intake of carbohydrates and is thought to act physiologically at times of energy depletion [Leibowitz 1989]. NPY levels are increased in animals that do not have leptin [Wilding et al. 1993] or functional leptin receptors 41 [Stephens et al. 1995]. In a study in which NPY-knockout mice were used it was shown that leptin remained effective on the suppression of feeding indicating that this action of letpin is mediated by pathways independent of NPY [Erickson et al. 1996]. In this study it was also suggested that NPY opposes the appetite-suppressing effects of leptin. It is known that (1) fasted animals have low leptin [Mizuno et al. 1996], (2) low leptin levels increase NPY [Schwartz et al. 1996a], and (3) high NPY inhibits GH [Okada et al. 1993] by increasing SRIH and by decreasing GHRH [McCann et al. 1989; Rettori et al. 1990]. In addition, NPY given centrally blunts leptin-induction of GH secretion [Carro et al. 1998]. It was also shown that the blunted GH that is observed in the fasted state occurs in conjunction with elevated NPY mRNA [Vuagnat et al. 1998]. When leptin was administered in this study, the NPY mRNA was reduced concomitant with normalized GH levels [Vuagnat et al. 1998]. It is therefore possible that, in addition to the other regulation pathways, leptin regulation of GH is mediated through NPY [Carro et al. 1997] or that leptin and NPY regulate GH secretion in parallel [Carro et al. 1998]. Objectives The regulation of GH by leptin and the effects on body homeostasis are the main topic of investigation for my dissertation. Most of the literature on GH and leptin interactions reviewed here was published after I developed my hypotheses. I hypothesized that (1) in the lean animal, circulating leptin stimulates the release of GH by acting directly at the level of the anterior pituitary and/or indirectly at the level of the hypothalamus, and (2) animals that are hyperleptinemic and therefore leptin resistant lose this ability to regulate GH. My work has been summed up by Scacchi et al.: Leptin, by favouring GH secretion, might reinforce its own biological effects, chiefly directed (as far as we presently know) at regulating the body fat content. 42 On the other side, the coexistence of high leptin and low GH serum levels in obesity fits in well with the concept of a leptin resistance in this condition. [1999, p.263] CHAPTER 2 GENERAL METHODS Animals Male Long-Evans rats (the strain from which the fa/fa rat was derived) were obtained from Harlan-Sprague Dawley (Indianapolis, IN) and housed individually under standard temperature and lighting conditions (12 hour light:dark cycle). Individual housing was required for individual food intake measurements. Water was available ad libitum at all times. All procedures using animals received prior approval by the Institution's Animal Care and Use Committee (IACUC). Diets The two test diets were obtained from PJ Noyes Company, Inc. (Lancaster, NH). The high-calorie, high-fat diet consisted of 20-23% fat and 3.7 kilocalories/gram (kcal/g). The high-calorie, low-fat diet consisted of 2-3% fat and 3.7 kcal/g. Normal rat chow #5001, obtained from Purina Mills, Inc. (Richmond, VA), consisted of 5% fat and 3.0 kcal/g. It should be noted that the low-fat diet had a calorie content equivalent to that of the high-fat diet, both of which were higher in calories than the control diet. Feeding, Pair-feeding, and Body Weight Measurements Feeding The animals were allowed an acclimation period of 5 days during which they became familiar with the new diets and daily handling. After acclimation, each of the diets was weighed (Mettler scale, PM200, Highstown, NJ) to the nearest 0.1 gram and administered 43 44 daily to each animal. Each subsequent day, the amount of diet remaining was recorded, including food that was dropped into or through the bottom of the cages. Daily intake was calculated by subtracting the amount of food remaining from the amount of food initially administered. Average daily intake was calculated per group. Pair-feeding After the acclimation period, the high fat and control rats were fed ad libitum daily. The low-fat rats were pair-fed the amount in grams ingested by the high-fat group the previous day. In this manner, the diets of the animals consuming the treatment diets differed only in the percentage of fat content, not in the number of calories. Body weight Body weights of the animals in all three groups were recorded daily to the nearest gram. The animals were weighed individually in a top loading scale (Taconic Farms, YG-700, Germantown, NY). Leptin Challenge Test The animals were food deprived for 24 hours prior to the test. The leptin challenge test consisted of a 30 mg/kg subcutaneous bolus of murine leptin (Amgen, Inc., Thousand Oaks, CA) given to half of the animals in each treatment group (leptin challenge). The other half of the animals in each group received a bolus of PBS for control. Food (Purina rat chow #5001, Purina Mills Inc., Richmond, VA) was returned to the animals one hour post-injection, allowing the animals time to recover following the injection. Food intake was then measured at 4 hours and again at 24 hours. 45 Alzet Osmotic Minipumps Alzet osmotic minipumps were used for the continuous administration of murine leptin (Amgen, Inc., Thousand Oaks, CA) or vehicle over a 2-week (Model 2ML2, Alza Scientific Products, Palo Alto, CA) or 4-week (Model 2ML4) period. Pump contents were delivered at a rate of 5 µg/hour or 2.5 µg/hour, respectively. Leptin or PBS (vehicle) was loaded into each pump under sterile conditions. Pumps were implanted subcutaneously under methoxyflurane anesthesia. Leptin was stable for the entire 4 week treatment period. For doses of leptin used, please refer to Chapters 5 and 6. Right Atrial Cannulation Anesthesia Sodium phenobarbital (65 mg/mL, 45-50 mg/kg, Veterinary Laboratories, Inc., Lenexa, KS) was administered intraperitoneally at a dose of 45-50 mg/kg to produce analgesia, anesthesia, and muscle relaxation. Atropine sulfate (0.1 mL of 1 mg/mL concentration, American Reagent Laboratories, Inc., Shirley, NY) was given intramuscularly in order to prevent the accumulation of fluid in the lungs. If the animals were incompletely anesthetized, methoxyflurane (Pitman-Moore, Inc., Mundelein, IL) was intermittently administered via a nose cone. Preparation All surgical equipment and cannula hardware were autoclaved or gas sterilized, as appropriate. The cranial (from between the ears to between the eyes) and neck areas (unilaterally from slightly caudal to the breastbone to the jaw) of the rats were shaved and disinfected with betadine and alcohol and the eyes were protected with lubricant. 46 Surgery In each rat, the right jugular vein was exposed and ligated to stop blood flow. A small cut was made in the jugular vein and the cannula was inserted and threaded toward the heart. Pulsation of the cannula indicated that it reached the heart. The cannula was then pulled back gently just until the pulsation ceased. At this location, the cannula was secured into position. The cannula was threaded subcutaneously and externalized at the base of the skull where it was secured with dental acrylic. Also fixed into the dental acrylic was a snap fastener to later attach to a snap when collecting blood. The cannula was filled with heparin to prevent coagulation. The neck incisions of the animals were closed with wound clips. Recovery Post-operatively, the animals were placed on heating pads in cages and were closely monitored until recovery from anesthesia. The animals were returned to their home cages until blood sampling. The wound clips were removed within 7-10 days of the surgery. Blood sampling cages The animals were each placed into the blood sampling cages two days prior to sampling to allow for acclimation to new surroundings. The cages are large wooden boxes into which wire mesh cages complete with bedding, food, and water were placed. Tubing was inserted into the top of the wooden cage, though a protective wire mesh, and attached to the cannula implanted into the rat, secured with the snap. Through this tubing, blood was collected without disturbing the rat, thus reducing confounding by stress hormones. Each cage has a light timer set to the same schedule as the standard housing facility and an exhaust system for the continual circulation of fresh air. This cage design was approved by the institution’s IACUC for temporary housing of rodents. 47 Blood Sampling and Tissue Collection Blood Collection from Cannulae Blood sampling occurred every 15 minutes for 6 hours from rats in the specialized cages described above. Before collection of each sample, 400 µL (the calculated amount of dead space, which includes heparinized saline used to keep the cannula patent) was withdrawn and saved. A sample of 300 µL was collected and dead space was returned immediately following the collection of the first sample. The blood was immediately centrifuged and the plasma was stored at -35°C until use in hormone and other assays. Red blood cells were resuspended in sterile heparinized saline and returned to the appropriate animals following collection of the subsequent blood sample and prior to the return of the dead space. This was done in order to prevent hypovolemia due to excessive sampling. This method was used to collect all remaining samples. Blood Collection from Tail Vein A scalpel was used to remove the tip of the tail from each unanesthetized rat and 1 mL of blood was collected into a tube. The tail abrasion required no treatment to stop the bleeding after the collection of blood. The blood was centrifuged at 3200 rpm for 30 minutes (Beckman Centrifuge Model J-6B, Fullerton, CA) and serum was frozen at -35ºC until use in hormone and other assays. Blood Collection via Cardiac Puncture Rats were anesthetized with methoxyflurane anesthesia and a 23-gauge needle was inserted into the heart to collect 1 mL of blood. The blood was centrifuged at 3200 rpm for 30 minutes (Beckman Centrifuge Model J-6B, Fullerton, CA) and serum was frozen at -35ºC until use in hormone and other assays. 48 Trunk Blood Collection The animals were sacrificed by decapitation and trunk blood was collected by holding the decapitated rat over a funnel and allowing blood to drain into a large glass test tube. Trunk blood was centrifuged at 3200 rpm for 30 minutes (Beckman Centrifuge Model J6B, Fullerton, CA) and serum was stored in the -35ºC freezer until use in hormone and other assays. Collection of Hypothalamus The animals were sacrificed by decapitation and the brain was dissected from the skull using sterile instruments and placed on ice. The hypothalamus was rapidly dissected away from the rest of the brain using a sharp razor. The hypothalamus was then rinsed, blotted dry, and weighed before being placed into a sterile tube and snap frozen in liquid nitrogen. The samples were frozen at -90ºC until reverse transcription and polymerase chain reaction (RT-PCR) and Western immunoblotting were initiated for leptin receptor. Pituitary and Organ Weights In the skull, the clear membrane over the sella turcica was broken with forceps. The posterior and intermediate lobes of the pituitary were removed and discarded. The anterior lobe of the pituitary was removed, rinsed, blotted dry, and weighed. From the body, the liver, testes, kidneys, heart, and adrenals were removed, rinsed, blotted dry, and weighed. Cell Culture GH1 Cells GH1 cells (American Type Culture Collection, Rockville, MD) are rat pituitary tumor cells that hypersecrete GH. Cells were grown to 70% confluency in F-12K media (ATCC, Rockville, MD) supplemented with 1% non-essential amino acids, 1% L- 49 glutamine, 1% nystatin (Gibco BRL, Life Technologies, Grand Island, NY), and either 10% horse serum and 2.5% fetal bovine serum (FBS, Gibco BRL, Life Technologies, Grand Island, NY) or 12.5% charcoal-stripped FBS (Hyclone, Logan, UT). Cells were incubated at 37ºC with 5% CO2. Media was changed 2-3 times weekly. GH1 cells were back-cultured weekly using standard trypsinization procedures to maintain the cell line. GH1 cells were used in passages 42-44. Plating Cells In general, cells were plated at 200,000 cells/well in 24-well plates in a volume of 1 mL/well. Cells were allowed time to adhere, usually 2-3 days, before experimentation. On the day of each experiment, medium was aspirated from each well and discarded. Control or leptin-supplemented (murine leptin, Amgen, Inc., Thousand Oaks, CA) medium was added to appropriate wells. Experiments were completed as described below. Five-Day Time -Course Control or leptin-supplemented (100nM) media was added to appropriate wells on each plate. Supernatant was collected each day for 5 days and frozen at -35ºC until GH RIA. Cells were collected and analyzed for DNA content for normalization of GH values and to control for potentially inconsistent plating densities. Media Experiment Cells were plated using media and supplements as described above, but with differences in serum content. Serum-free media or media supplemented with either 10% horse serum and 2.5% FBS or with 12.5% charcoal stripped FBS were utilized. The rationale for these differences in serum supplementation are explored in Chapter 4. 50 Collecting RNA from GH1 Cells RNA from GH1 cells was collected for RT-PCR amplification of leptin receptor mRNA. Media was aspirated from the flask and discarded. Trizol (Gibco BRL, Life Technologies, Grand Island, NY), a reagent designed for the isolation of total RNA from tissues and cells, was added to the flask (1 mL/10 cm2) and the cells and Trizol were triturated. Trizol and cells were collected and RNA was extracted as described below. RT-PCR was performed as described below. Radioimmunoassays GH Iodination 125 I-Na (1 mCi, Amersham Life Science, Inc., Arlington Heights, IL) was added to a 10 µL aliquot of 1 mg/mL rGH-I-6 (NIADDK, NIH National Pituitary Agency, Bethesda, MD) using a lead shielded syringe. Chloramine T (25 µL of 1.5 mg/mL, Sigma Chemical Co., St. Louis, MO) was added to initiate the reaction and 50 µL sodium metabisulfite (2.4 mg/mL, Fisher Scientific, Pittsburg, PA) was added 55 seconds later to terminate the reaction. Bovine serum albumin (100 µL of 100 mg/mL RIA grade, Sigma Chemical Co., St. Louis, MO) was added to coat the column. The entire solution was placed on a Sephadex G-75 (Sigma Chemical Co., St. Louis, MO) or Bio-Gel P-60 (BioRad Laboratories, Richmond, CA) column for separation. Each sample was counted on the Apex Automatic Gamma Counter (ICN Micromedic Systems Model 28023, Huntsville, AL with RIA AID software, Robert Maciel Associates, Inc., Arlington, MA) and the fraction with the highest level of radioactivity was saved and diluted for use in GH radioimmunoassays (RIAs). 51 Growth Hormone RIA GH was measured in duplicate (cell culture media) or triplicate (plasma) using materials supplied by Dr. A. F. Parlow and the National Hormone and Pituitary Program (NIADDK, Baltimore, MD). Values were expressed in ng/mL in terms of the NIADDK reference preparation rat GH-RP-2. Plasma collected from hypophysectomized rats was added to the standard curve in the rat plasma assays to correct for non-specific binding. Hypophysectomized rats have had their anterior pituitaries removed. The plasma, therefore, lacks GH but has the GH binding proteins. Monkey-anti-rat GH primary antibody, diluted 1:70,000, and labeled GH, diluted to approximately 12,000 counts/100 µL, were added to the assay and incubated at room temperature for 3 days. On day 4, goat-anti-monkey secondary antibody, diluted 1:30, was added. Normal monkey serum (1:200) was added with the secondary antibody to reduce non-specific binding. The assay was incubated at room temperature for 1 day. On day 5, all tubes except total counts were centrifuged for 30 minutes (3200 rpm, Beckman Centrifuge Model J-6B, Fullerton, CA) at 4°C. Supernatant was removed with a vacuum aspirator and pellets were counted on the Apex Automatic Gamma Counter (ICN Micromedic Systems Model 28023, Huntsville, AL with RIA AID software, Robert Maciel Associates, Inc., Arlington, MA) for 1 minute. The GH RIA has an assay sensitivity of 1 ng/mL and a range of detection from 1 ng/mL to 320 ng/mL. IGF Iodination 125 I-Na (1 mCi, Amersham Life Science, Inc., Arlington Heights, IL) was added to a 10 µL aliquot 0.25 mg/mL IGF-1 iodination preparation (BACHEM Bioscience, Inc., King of Prussia, PA) using a lead shielded syringe. Chloramine T (10 µL of 1.0 mg/mL, Sigma Chemical Co., St. Louis, MO) was added to initiate the reaction. After 45 52 seconds, 200 µL of 100 mg/mL bovine serum albumin (RIA grade, Sigma Chemical Co., St. Louis, MO) was added. The entire solution was placed on a Sephadex G-75 column (Sigma Chemical Co., St. Louis, MO) for separation. Each sample was counted on the Apex Automatic Gamma Counter (ICN Micromedic Systems Model 28023, Huntsville, AL with RIA AID software, Robert Maciel Associates, Inc., Arlington, MA) and the fraction with the highest level of radioactivity was diluted and saved for use in IGF RIAs. IGF RIA IGF-1 was extracted from serum by the acid/ethanol procedure. IGF-1 was then measured by RIA; plasma samples were measured in duplicate for IGF. Values were expressed in ng/mL in terms of the BACHEM IGF reference preparation (BACHEM Bioscience, Inc, King of Prussia, PA). Rabbit-anti-rat IGF primary antibody, diluted 1:3,000, and labeled IGF, diluted to approximately 12,000 counts/100 µL, were added to the assay and incubated at 4°C for 2 days. On day 3, goat-anti-rabbit secondary antibody, diluted 1:20, was added. Normal rabbit serum (1:50) was added with the secondary antibody to reduce non-specific binding. The assay was incubated at 4°C for 1 day. On day 4, all tubes except total counts were centrifuged for 30 minutes (3200 rpm, Beckman Centrifuge Model J-6B, Fullerton, CA) at 4°C. Supernatant was removed with a vacuum aspirator and pellets were counted on the Apex Automatic Gamma Counter (ICN Micromedic Systems Model 28023, Huntsville, AL with RIA AID software, Robert Maciel Associates, Inc., Arlington, MA) for 1 minute. The IGF RIA has an assay sensitivity of 0.1 ng/mL and a range of detection of 0.1 ng/mL to 20 ng/mL. Leptin RIA Leptin was analyzed with a rat leptin RIA kit (Linco Research, Inc., St. Charles, MO) which measures both rat and mouse leptin with an assay sensitivity of 0.5 ng/mL and a 53 range of detection from 0.5 ng/mL to 50 ng/mL. Briefly, samples and standards were aliquoted in duplicate and primary guinea pig antibody, raised against highly purified rat leptin, was added. The antibody is 100% cross-reactive with rat and mouse leptin and <2% reactive with human leptin. The tubes were incubated overnight at room temperature. On day 2, 125I-leptin was added and the tubes were incubated overnight at room temperature. On day 3, cold precipitating reagent was added and the tubes were incubated at 4°C for 20 minutes. All tubes except total counts were centrifuged for 40 minutes (3200 rpm, Beckman Centrifuge Model J-6B, Fullerton, CA) at 4°C. Supernatant was removed with a vacuum aspirator and pellets were counted on the Apex Automatic Gamma Counter (ICN Micromedic Systems Model 28023, Huntsville, AL with RIA AID software, Robert Maciel Associates, Inc., Arlington, MA) for 1 minute. Insulin RIA Insulin was measured with a rat insulin RIA kit (Linco Research, Inc., St. Charles, MO) with an assay sensitivity of 0.1 ng/mL and a range of detection from 0.1 ng/mL to 10 ng/mL. Briefly, samples and standards were aliquoted in duplicate and primary guinea pig antibody, raised highly purified rat insulin, and 125I-insulin were added. The tubes were incubated overnight at 4°C. On day 2, cold precipitating reagent was added and the tubes were incubated at 4°C for 20 minutes. All tubes except total counts were centrifuged for 40 minutes (3200 rpm, Beckman Centrifuge Model J-6B, Fullerton, CA) at 4°C. Supernatant was removed with a vacuum aspirator and pellets were counted on the Apex Automatic Gamma Counter (ICN Micromedic Systems Model 28023, Huntsville, AL with RIA AID software, Robert Maciel Associates, Inc., Arlington, MA) for 1 minute. The assay is 100% specific for rat, mouse, hamster, human, porcine, and ovine insulin. 54 Glucose and Triglyceride Assays Glucose was measured in serum or plasma using a YSI 1500 Sidekick Glucose Analyzer (Yellow Springs Instrument Company, Inc., Yellow Springs, OH). This instrument utilizes the glucose oxidase method and the results are linear from 0-800 mg/dL. A 180 mg/dL standard was used. Triglycerides were measured using a kit from Sigma (Sigma Diagnostics, St. Louis, MO). Triglycerides are hydrolyzed and glycerol is measured with a qualitative enzymatic method. A 250 mg.dL standard was used and the assay is linear to a triglyceride level of 1000 mg/dL. DNA Assay DNA of GH1 cells was assayed to normalize GH values. After collection of cell culture media from the 48-well plates (see Cell Culture section), high salt DNA assay buffer (0.05M sodium phosphate, 2.0M NaCl, 2 mM EDTA, pH 7.4) was added to the cells in each well and incubated overnight. The samples were collected and sonicated prior to DNA measurement. The DNA standard curve was derived from calf thymus (Sigma Chemical Co., St. Louis, MO) and Hoescht florescence emission dye (Molecular Probes, Eugene, OR) was used. Hoescht dye binds to intact double stranded DNA. Standards and samples were aliquoted into opaque (white or black) 96-well plates in triplicate. Hoescht dye (1 µg/mL) was added and the plate was read on a Molecular Devices Type 374 Fluorometer (Labsystems, Finland) using SOFTmax PRO Version 1.3.2 software for Macintosh at excitation wavelength 320 nm and emission wavelength 520 nm. 55 RNA extraction, RT-PCR, Southern Blot RNA Extraction RNA was extracted from hypothalamus and GH1 cells using the phenol-chloroform method with isopropyl alcohol precipitation. Hypothalami were homogenized and GH1 cells were collected in Trizol reagent (Gibco BRL, Life Technologies, Grand Island, NY). At the end of the procedure, the pellet was dried and resuspended in RNAse-free water. Absorbances were read on a spectrophotometer (Model DU-64, Beckman, Fullerton, CA) to obtain an OD260:OD280 value. OD260:OD280 of 1.7 to 2.1 indicates a clean RNA extraction. RT-PCR for Leptin Receptor RT-PCR for leptin receptor mRNA was optimized using the sense and antisense primers designed by Zamorano et al. [1997]. The sense and antisense primers were 5’ATGACGCAGTGTACTGCTG and 5’-GTGGCGAGTCAAGTGAACCT, respectively and amplified a 357-base pair fragment of the long form of the leptin receptor. The primers were designed using a homologous region in mouse and human leptin receptor cDNA to amplify the extracellular domain (bp 1274-1630) of the leptin receptor in the rat (95% homologous with rat). Optimal conditions were identified as 1 mM MgCl 2, 64°C annealing temperature (Figure 2-1), and 30 cycles (Figure 2-2) following an initial denaturation phase and followed by a final elongation phase. No plot was made of density vs. temperature, MgCl 2, or cycle number. The determination of optimal conditions was made by rough estimation. Negative controls, including a sample lacking 56 the reverse transcription enzyme and a sample lacking RNA, were included (data not shown). Figure 2-1: MgCl 2 (mM) and temperature (°° C) optimization Lane 1 is the 100 base pair (bp) DNA ladder; lane 2 is 0.5 mM at 63°C; lane 3 is 1.0 mM at 63°C; lane 4 is 1.5 mM at 63°C; lane 5 is 2.0 mM at 63°C; lane 6 is 0.5 mM at 64°C; and lane 7 is 1.0 mM at 64°C. Based on the results of this experiment, the conditions used for the subsequent studies were 1 mM MgCl 2 at 64°C. Figure 2-2: Cycle optimization Lane 1 is the mass DNA ladder; lane 2 is 25 cycles; lane 3 is 30 cycles; lane 4 is 35 cycles; and lane 5 is 40 cycles. Based on the results of this experiment, 30 cycles were used in subsequent experiments. Southern Blot and Normalization The identity of RT-PCR product (Figure 2-3) was confirmed using the internal probe 5’TGCAGCTGAGGTATCACAGG in Southern blot analysis (ECL, Amersham Pharmacia Biotech, Piscataway, NJ). The inconsistencies in lanes 2 and 3 of the southern blot are due to relative differences of RNA added to the initial PCR. The amount of leptin receptor mRNA was normalized with the housekeeping gene cyclophilin. Cyclophilin did not appear to be affected by the treatment parameters. The cyclophilin sense and antisense primers used were 5'-GGGAAGGTGAAAGAAGGCAT and 5'GAGAGCAGAGATTACAGGGT, respectively and amplified a 210-base pair fragment [Zamorano et al. 1997]. Cyclophilin conditions were optimized and found to be the same as were used for the leptin receptor but half the concentration of RNA was added to the 57 reaction to prevent saturation of cyclophilin. A representative photograph of RT-PCR is shown in Figure 2-4. Figure 2-3: Southern blot of leptin receptor Lane 1 is hypothalamus; lanes 2 and 3 are GH1 cells; and lanes 4 and 5 are RC cells. GH1 and RC cells are rat pituitary tumor cell lines that hypersecrete and hyposecrete GH, respectively. Figure 2-4: Representative RT-PCR from hypothalamus samples Lane 1 is the mass DNA ladder; lanes 2, 4, 6, and 8 are leptin receptor mRNA; and lanes 3, 5, 7, and 9 are corresponding cyclophilin mRNA. Protein Extraction and Western Immunoblotting Protein Extraction Trizol: Protein was extracted from the same samples from which RNA was extracted using Trizol reagent (Gibco BRL, Life Technologies, Grand Island, NY). After addition of phenol/chloroform to isolate RNA, RNA was in the aqueous phase, DNA was in the interphase, and protein remained in the phenol/chloroform phase. From the phenol/chloroform phase, protein was precipitated with isopropanol and washed in a solution containing guanidine hydrochloride. The samples were centrifuged at 7,500 g at 4°C (Beckman Centrifuge Model J2-21, Fullerton, CA) and the pellet was dried and resuspended in 1% SDS. However, the Trizol reagent does not contain a detergent to efficiently break cell membranes and release membrane-bound proteins, so additional samples were extracted in cell lysis buffer (see below) and comparisons were made in Figures 2-5 and 2-6. 58 Cell Lysis Buffer: Some hypothalami did not require RNA extraction prior to protein extraction, and therefore were not extracted using the Trizol method. Instead, hypothalami were homogenized in a phosphate buffer (g tissue x 10 mL buffer), pH 7.4, containing 5 mM EDTA, 5 mM EGTA, 50 mM NaCl, 5 mM sodium pyrophosphate decahydrate, 2% Triton X-100, 0.5% SDS (all from Fisher Scientific, Fair Lawn, NJ), 1 mM orthovanadate, 0.1 mM PMSF (both from Sigma Chemical Co., St. Louis, MO), 10 ug/mL leupeptin, 1.0 ug/mL pepstatin, 10 ug/mL aprotinin (all three from Calbiochem, La Jolla, CA), and 50 mM NaF (Fisher Scientific, Fair Lawn, NJ) using a Brinkman polytron homogenizer (Brinkman Instruments, Inc., Palo Alto, CA). After homogenization, samples were kept on ice and vortexed periodically. After 1 hour, samples were centrifuged (Micro Centrifuge Model 235C, Fisher Scientific, Fair Lawn, NJ) for 30 minutes and supernatant was frozen at -20°C until further use. Figure 2-5: Trizol extraction Lane 1 is the molecular weight marker; lanes 2-6 are hypothalamic samples extracted by the trizol method. Leptin receptor (long form) should be near the 126 kDa marker. Figure 2-6: Cell lysis buffer extraction Lane 1 is the molecular weight marker; lanes 2-9 are hypothalamic samples extracted by the cell lysis buffer protocol. Leptin receptor (long form) is near the 126 kDa marker. Micro BCA Protein Assay Micro BCA protein assay is used to measure protein when the samples are suspended in reagents that interfere with the Coomassie blue of the Bradford protein assay, such as SDS. A standard curve ranging from 0.063 mg/mL to 1.0 mg/mL was made by serial dilution in 1% SDS starting with 2 mg/mL albumin standard (Peirce, Rockford, IL). A 59 reference standard (0.0 mg/mL) was also used. Each standard was pipetted into a colorless 96-well plate in triplicate. Samples were added in duplicate. If dilutions were required, 1% SDS was used as the diluent. A working reagent was prepared by mixing reagents MA, MB, and MC (Micro BCA reagents, Pierce, Rockford, IL) at a ratio of 25:24:1. Working reagent was added to standards and samples in each well. The plate incubated at 37°C for 30 minutes. The plate was then read on the SLT 400 AC Plate Reader (SLT Lab Instruments, Salzburg, Austria) using dual wavelength (575 nm and 690 nm) with 30 second shake and four parameter standard curve fit. The accompanying software was DeltaSoft II for Macintosh (BioMetallics, Inc., Princeton, NJ). Western Immunoblot Analysis for Leptin Receptor Separation and transfer: The 10 µL protein sample or molecular weight marker (Kaleidoscope, BioRad Laboratories, Hercules, CA) was combined with 5 µL loading buffer (0.06 M Tris, 2% SDS, 10% glycerol, 0.025% bromphenol blue, and 5% 2mercaptoethanol), boiled, and loaded onto the pre-cast polyacrylamide-SDS Ready Gel (BioRad Laboratories, Richmond, CA). For protein loading optimization, see Figure 2-7. Different volumes of protein were evaluated, 2-9 µL. The best results were seen at a volume of 9 µL, but the bands were still faint. In all subsequent Westerns, the maximum amount of protein (10 µL) was loaded. After the Western was completed, µg protein loaded was calculated. No negative controls were utilized. After loading onto the gel, the samples were separated by running the gel in a gel electrophoresis apparatus (MiniPROTEAN II Cell, BioRad Laboratories, Hercules, CA) at 125 V for approximately 70 minutes. Samples were transferred to a nitrocellulose membrane (Trans-Blot Transfer Medium, 0.45 um, 7 x 8.4 cm; BioRad Laboratories, Hercules, CA) at 100 V for 1 hour using a BioRad Trans Blot Cell apparatus (Hercules, CA). 60 Immunoblotting and detection: The membrane was blocked for 2 hours in 5% nonfat dry milk in Tris buffered saline with 0.05% Tween (T-TBS) and then incubated with primary antibody overnight at 4°C. The primary antibody used was rabbit-anti-rat leptin receptor (long-form) produced to the 18 amino acids near the C-terminus of mouse ObRb (Alpha Diagnostic International, San Antonio, TX). The primary antibody was diluted 1:500 in blocking buffer. After 3 10-minute washes with T-TBS, the membrane was incubated with the secondary antibody (anti-rabbit IgG-HRP, Santa Cruz Biotechnology, Santa Cruz, CA) at a dilution of 1:1,000 for 1 hour. Following another washing step, the blot was exposed to 5 mL of each solution (simultaneously) of SuperSignal Chemiluminescent Substrate for Western blotting (Pierce, Rockford, IL) for 10 minutes. The blot was then exposed to radiographic film (Hyperfilm ECL, Amersham Life Science, Buckinghamshire, England) for 10 minutes. The film was developed in a Konica Medical Film Processor (QX-70). Optimization for the antibody concentrations is shown in Figure 2-8. The best combination of primary and secondary antibody shown in the figure is with each at a dilution of 1:1000, but the resutling bands were faint. A subsequent test used a more concentrated primary antibody (1:500) with the same concentration of secondary antibody (1:1000), with better results (data not shown). These were the conditions used for all subsequent Western analyses. Analysis: A pooled reference sample was included in each gel to correct for inter-gel variation. The optical density of the bands was measured using an imaging densitometer (BioRad Model GS-670, Hercules, CA) and Molecular Analyst Software (version 1.2, Hercules, CA). The results were normalized according to the amount of protein loaded (µg) and expressed as a ratio of sample to inter-gel control. 61 Figure 2-7: Protein loading optimization Lane 1 is the molecular weight marker; lane 2 is 2 µL; lane 3 is 3 µL; lane 4 is 4 µL; lane 5 is 5 µL; lane 6 is 6 µL; lane 7 is 7 µL; lane 8 is 8 µL; and lane 9 is 9 µL of a 2.74 µg/µL sample. In subsequent experiments, 10 µL of each protein sample was used. Figure 2-8: Primary antibody concentration optimization Lanes 1, 3, 5, and 7 are the molecular weight markers; lane 2 is 1:1000; lane 4 is 1:1500; lane 6 is 1:2000; and lane 8 is 1:2500. In lanes 2, 4, 6, and 8, secondary antibody concentration was 1:1000. In subsequent experiments, 1:500 primary antibody and 1:1000 secondary antibody concentrations were used. Each leptin receptor protein measured with this Western protocol gave bands of multiple molecular weights. No pre-immune or pre-absorption samples were run, so the specificity of the bands is uncertain. The molecular weight of the largest band is what was expected for the leptin receptor, so it can be assumed to be specific. The lower molecular weight bands have been seen in studies using different antibodies [Boes et al. 1999], so these are possibly specific as well. CHAPTER 3 CIRCULATING GROWTH HORMONE LEVELS ARE ELEVATED IN RATS FED A HIGH-FAT DIET Introduction It is well known that the secretion of growth hormone (GH) from the anterior pituitary gland is episodic. In the past, the rat has been the most widely studied model for investigation of basic mechanisms that underlie the expression of the GH secretory pattern in man because of numerous similarities in the GH secretory axes between man and rats. The regulation of GH secretion is under direct hypothalamic control and is mediated by the release of a stimulatory neuropeptide, growth hormone releasing hormone (GHRH) and an inhibitory neuropeptide, somatotropin release inhibiting hormone (SRIH), also called somatostatin [Martin and Millard 1986; Millard 1989]. In addition to hypothalamic control, metabolic control of GH secretion also shares similarities in these species. It is clear that circulating GH is dramatically attenuated in both obese animals [Veldhuis et al. 1991] and obese humans [Williams et al. 1984] and that this may be directly related to blood-borne factors and their influence on hypothalamic neurons involved in GH regulation. There are other metabolic controllers of GH, however, that have the opposite effects between man and rodent species, such as exercise and fasting. One possible example of a circulating GH-regulating factor is the obese gene product, leptin. Leptin is a protein hormone secreted by adipocytes in proportion to body fat that acts in the brain to help regulate body weight. When there is insufficient leptin or 62 63 impaired leptin recognition by the brain, obesity develops. Circulating leptin influences and is influenced by metabolic states such as obesity, exercise, and fasting [Mizuno et al. 1996; Ahren et al. 1997]. It is therefore logical to assume that body weight and consequently leptin may influence GH secretion. An additional argument for this is that leptin receptors are highly abundant in the arcuate nucleus of the hypothalamus (ARC) [Mercer et al. 1996], which is also the location of GHRH-producing neurons [Martin 1973]. Leptin, being secreted in proportion to body fat, is higher in the blood of animals with greater fat deposition. Consequently, it tends to be elevated in the blood of animals that ingest a diet high in fat [Campfield et al. 1995; Frederich et al. 1995; Masuzaki et al. 1995; Van Heek et al. 1997; Widdowson et al. 1997]. The typical human diet generally consists of fat in excess of 20% [Wang et al. 1998a], which is much greater that the laboratory rodent diet that is approximately 5% fat (Purina Mills rodent chow). We proposed that by placing young rats on a high-fat diet we would elevate circulating leptin to levels, which would subsequently cause an elevation of circulating GH, assuming that the length of time the rats were on the diets and the levels to which leptin was raised were not sufficient to cause obesity/resistance. The results of the present study confirm this hypothesis. Methods Animals Twenty-five male Long-Evans rats (300-325 g) were obtained from Harlan-Sprague Dawley and housed individually under standard temperature and lighting conditions. The animals were divided into three groups each receiving different diets: 9 rats received a 64 high-fat diet (23% fat, 3.7 kcal/g, PJ Noyes Company, Inc., Lancaster, NH), 8 received an isocaloric, low-fat diet (2% fat, 3.7 kcal/g, PJ Noyes Company, Inc., Lancaster, NH) which was high in carbohydrates, and 8 controls received normal rat chow (5% fat, 3.0 kcal/g, Purina Mills, Inc., Richmond, VA). The control and high-fat rats were fed ad libitum. The low-fat rats were pair-fed the amount of food in grams consumed by the high-fat rats the previous day. Water was available to all groups ad libitum. Body weight and food intake were measured daily in diet-treated rats and weekly in controls for the duration of the study. Cannulation Surgery, Blood Sampling, and Tissue Collection After four weeks on the various diets, the rats were implanted with right atrial cannulae under pentobarbital anesthesia (45-50 mg/kg intraperitoneally). The cannulae were externalized at the base of the skull and secured with dental acrylic. One week after surgery, 300-400 µL blood samples were collected at 15-minute intervals for a total of 6 hours, beginning between 8 and 9 AM for each rat. Immediately after each 15-minute sample collection, the blood was centrifuged and the plasma was stored at -35°C until hormone assays. Red blood cells were resuspended in sterile heparinized saline and returned to each respective animal following collection of the subsequent blood sample. This was done in order to prevent hypovolemia due to excessive sampling. After the completion of sampling, the animals were sacrificed and trunk blood was collected for analysis of insulin-like growth factor-1 (IGF-1) and leptin. Hypothalami were removed and frozen at -90°C until reverse transcription and polymerase chain reaction (RT-PCR) studies were initiated for leptin receptor. 65 Radioimmunoassays Plasma samples were measured in triplicate for GH using materials supplied by Dr. A. F. Parlow and the National Hormone and Pituitary Program (NIADDK, Baltimore, MD), as described [Millard et al. 1981]. Values were expressed in ng/mL in terms of the NIADDK reference preparation rat GH-RP-6. The GH RIA has an assay sensitivity of 1 ng/mL and a range of detection of 1 ng/mL to 320 ng/mL. Plasma samples were measured in duplicate for IGF-1. IGF-1 was extracted from trunk blood by the acid/ethanol procedure and measured by RIA as previously described [Grant et al. 1986]. The IGF RIA has an assay sensitivity of 0.1 ng/mL and a range of detection of 0.1 ng/mL to 20 ng/mL. Leptin was measured with a RIA kit (Linco Research, Inc., St. Charles, MO) which measures both rat and mouse leptin with an assay sensitivity of 0.5 ng/mL and a range of detection from 0.5 ng/mL to 50 ng/mL. RT-PCR for Leptin Receptor RT-PCR for leptin receptor mRNA was optimized using the sense and antisense primers from Zamorano et al. [1997]. The sense and antisense primers were 5’ATGACGCAGTGTACTGCTG and 5’-GTGGCGAGTCAAGTGAACCT, respectively and amplified a 357-base pair fragment of the long form of the leptin receptor. Optimal conditions were identified as 1 mM MgCl 2, 64°C annealing temperature, and 30 cycles following an initial denaturation phase and followed by a final elongation phase. The identity of RT-PCR product was confirmed using the internal probe 5’TGCAGCTGAGGTATCACAGG in Southern blot analysis (ECL, Amersham Pharmacia Biotech, Piscataway, NJ). The amount of leptin receptor mRNA was normalized with the housekeeping gene cyclophilin. The cyclophilin sense and antisense primers used were 5'-GGGAAGGTGAAAGAAGGCAT and 5'-GAGAGCAGAGATTACAGGGT, 66 respectively and amplified a 210-base pair fragment [Zamorano et al. 1997]. The same conditions were used for cyclophilin as were used for the leptin receptor. Statistics Daily and weekly body weight and food intake measurements were compared by twoway repeated measures ANOVA with Student-Newman-Keuls Multiple Comparisons. Growth hormone profiles were evaluated by Cluster Analysis [Veldhuis and Johnson 1986], which analyzes area under the curve, peak height and width, and valley nadir and width. Comparisons of IGF-1, leptin, and leptin receptor mRNA were by one-way ANOVA followed by Tukey Comparisons. Values were significant using p<0.05 unless otherwise indicated. Results Body Weight Although rats fed the low-fat diet did not lose weight during the study, they did not gain as much weight as rats fed either the high-fat or control diets (Figure 3-1). There were significant differences in body weights between low- and high-fat rats beginning on day 10 and remaining for most of the duration of the study and between the low-fat and control rats on days 10-14 and 18-29. Rats fed the high-fat diet gained significantly more weight than controls beginning at day 18 and remaining for most of the duration of the study. Food Intake Rats fed the high-fat diet consumed significantly less chow in grams of food than rats fed the control diet beginning within the first 5 days of the study and remaining for the duration (Figure 3-2). No comparisons for animals fed the low-fat diet are reported 67 versus either of the other two groups because these rats were pair-fed (in grams of food) to the high-fat rats. When the data are shown as grams of fat consumed for each respective diet (Figure 3-2), the high-fat rats consumed significantly more throughout the study compared to both controls and low-fat rats. In addition, control rats consumed significantly more fat than low-fat rats for the duration of the study. Control rats consumed more kcal than rats fed the special diets. Leptin At the end of the study, leptin levels were significantly elevated in the rats fed the highfat diet compared to those fed either of the other two diets (Figure 3-3). Although it appeared that rats fed normal chow had lower leptin values than low-fat rats, the results were not significant. Leptin Receptor mRNA There were no significant differences in leptin receptor mRNA in hypothalamus in response to any of the different diets (Figure 3-4). IGF At the end of the study, IGF-1 levels were significantly elevated in the rats fed the highfat diet compared to those fed either of the other two diets (Figure 3-5). Growth Hormone Profile Growth hormone pulses and troughs were analyzed using the Cluster Program [Veldhuis and Johnson 1986] among the diet-treated and control groups (Table 3-1). It must be pointed out, however, that each group only averaged two or three values due to sampling difficulties. Analyses were completed and demonstrated that, overall, the high-fat rats had the highest total and mean GH under-the-curve values while the low-fat rats had the lowest (p<0.10). This was most likely due to peak height and valley width. High-fat rats 68 had significantly higher peaks while low-fat rats had significantly lower peaks overall compared to control (p<0.05). Similarly, the valley nadirs were higher in high-fat rats and lower in low-fat rats, but these results were not significant. Control rats were in the middle of the range for both peak height and valley nadir. As expected, because of elevated pulse amplitudes, the high-fat rats spent less time in the GH trough periods (valley width, p=0.01) compared to the other groups. Peak width, measured in minutes, was not significantly different among the groups. Representative profiles for the high-fat (panel A), control (panel B), and low-fat rats (panel C) are given in Figure 3-6. Figure 3-1: Body Weight There were significant differences in body weights between low- and high-fat rats (n=9) beginning on day 10 and remaining for most of the duration of the study and between the low-fat (n=8) and control rats (n=8) on days 10-14 and 18-29 (2-way repeated measures ANOVA, p<0.05). Rats fed the high-fat diet gained significantly more weight than controls beginning at day 18 and remaining for most of the duration of the study. 69 Figure 3-2: Food Intake When measured in grams (g food), it was shown that rats fed the high-fat diet (n=9) consumed significantly less chow than rats fed the control diet (n=8) beginning within the first 5 days of the study and remaining for the duration (2-way repeated measures ANOVA, p<0.05, panel A). No comparisons for animals fed the low-fat diet (n=8) are reported versus either of the other two groups because these rats were pair-fed in grams of food to the high-fat rats. When measured in grams of fat consumed (panel B), the high-fat rats consumed significantly more than the control rats, which in turn consumed significantly more than the lowfat rats (2-way repeated measures ANOVA, p<0.05). When measured in kcal (panel C), normal chow rats ate more for most of the study, but the values were nearly normalized. Figure 3-3: Leptin Levels At the end of the study, leptin levels were significantly elevated (1-way ANOVA, p<0.05) in the rats fed the high-fat diet (n=8) compared to those fed either of the other two diets (n=6 each). 70 Figure 3-4: Ob-Rb mRNA There were no significant differences (1-way ANOVA, p<0.05) leptin receptor mRNA in hypothalamus due to any of the differenct diets (n=6 for each group). Leptin receptor was normalized using the housekeeping gene cyclophilin. Figure 3-5: IGF At the end of the study, IGF-1 levels were significantly elevated (1-way ANOVA, p<0.05) in the rats fed the high-fat diet (n=8) compared to those fed either of the other two diets (n=6 each). 71 Table 3-1: Growth Hormone Profile of Diet-Treated and Control Rats Figure 3-6: GH Profile Representative GH profiles of high-fat (panel A), control (panel B), and low-fat rats (panel C). 72 Discussion In the mammalian endocrine system, a variety of hormones and their hormonal and metabolic regulators are intricately intertwined, both centrally and in the periphery. GH is one such hormone. GH stimulates IGF-1, and separately or in concert, GH and IGF-1 regulate body composition. In man, GH secretion is enhanced in diabetes [Glass et al. 1981] and virtually all forms of stress. Both exercise and fasting also increase GH [Martin and Millard 1986; Borst et al. 1994] while its secretion is attenuated in obesity. In rats, insulin and corticosterone have a negative effect on circulating GH levels [Tannenbaum et al. 1981], as do exercise and fasting. Similar to man, GH secretion is attenuated in obesity in rats [Martin et al. 1983]. The differential regulation of GH between rodents and man is largely unexplained. Leptin is a hormone that is regulated by IGF-1 [Bianda et al. 1997] and GH [Florkowski et al. 1996]. Upon GH replacement in GH-deficient subjects, body fat decreased resulting in a decrease of elevated leptin [Florkowski et al. 1996]. In another study, leptin levels were found to be elevated in GH-deficient adults and were normalized after 1 year of GH therapy [Fisker et al. 1997]. Although the authors claimed there was no association between the two hormones and that the effects were most likely due to the decrease in body fat which occurred as a result of the lipolytic effects of GH, there certainly is evidence that there is at least an indirect relationship. In addition, leptin is required for maximal blood GH levels [Carro et al. 1997]. Carro et al. [1997] demonstrated that leptin antiserum decreased GH amplitude and nadir in rats, suggesting that normal levels of leptin are required for normal GH secretion. They also showed that, in fasted animals with low leptin and low GH, administration of exogenous leptin normalized GH secretion. 73 GH has many important roles in the body, including lipolysis. Several investigators have shown GH receptors on adipocytes [Fagin et al. 1980; Vikman et al. 1991]. In obese animals, GH burst amplitudes are attenuated [Veldhuis et al. 1991]. This reduction of GH in obesity prevents GH-induced lipolysis, perhaps further contributing to obesity. This is another example of GH-leptin interaction. In the current study, rats consuming the high-fat diet gained more weight than controls. The weight gain of the high-fat rats occurred in spite of a food intake in grams and kilocalories significantly lower than that of controls. These results seem contradictory but are reasonable when the fat content of each diet is taken into effect. The grams of fat consumed by the high-fat group exceeded that of the control group, which in turn exceeded that of the low-fat group. Furthermore, rats that ate the low-fat diet gained less weight than controls, explained by the differences in overall fat consumption. Corresponding to the body weight data, leptin levels were significantly elevated in the high-fat rats versus the other two groups. This was expected and is probably due to elevated fat mass in these rats. The levels to which leptin was elevated remained within the a physiological range, indicating that the animals were not obese and probably were not leptin resistanct. In addition to elevated leptin, IGF-1 levels were significantly elevated in the high-fat rats. It has been shown that, in rats, IGF-1 is present in the blood at relatively constant concentrations dependent on GH secretory status [Donaghue et al. 1990]. IGF-1, therefore, is a good measure of overall GH secretion. Although IGF-1 is regulated by factors other than GH, it is often used clinically to detect GH secretory 74 problems. The elevation of IGF-1 in response to the high-fat diet may therefore suggest that GH was also elevated in this group. A growth hormone profile consisting of samples collected at 15-minute intervals for 6 hours was established for each of the three groups. Unfortunately, due to sampling difficulties, the number of animals in each group is insufficient to draw conclusions with any degree of confidence. However, the results in Table 3-1 strongly suggest that rats fed the high-fat diet secreted more GH and rats fed the low fat diet secreted less GH when compared to control rats. Low-fat rats were pair-fed and thus were probably still hungry when their food supply ran out, however it is unknown how much more food the low-fat rats would have consumed had they been fed ad libitum. Thus, the implied chronic stress experienced by these rats potentially due to being in a state of hunger may confound GH data. The results of the IGF-1 assays and GH profiles, taken together, suggest that ingestion of the high-fat diet and the subsequently elevated plasma leptin levels resulted in increased circulating GH values. Although correlation does not imply causation, it is possible that the physiological elevations in leptin levels stimulated the release of GH, either directly or indirectly. The literature suggests that leptin is required for normal GH release [Carro et al. 1997]. A natural feedback loop involving leptin and GH seems evident. Consider this greatly simplified physiological situation: when an animal begins to eat more and gain weight, resulting in hypertrohy and/or hyperplasia of fat cells, more leptin is produced. This elevation of leptin is sensed by the hypothalamus which decreases feeding and stimulates metabolism and perhaps directly by the pituitary to enhance GH secretion. GH 75 then acts directly on adipocytes [Fagin et al. 1980; Vikman et al. 1991] to reduce their size and number, and this with the decreased food intake result in less fat and decreased or normalized leptin production. When this fall in leptin is sensed by the hypothalamus, food intake is again increased and GH secretion is reduced in an effort to prevent lipolysis until normal fat mass has been attained. In the case presented in the current study, the animals are given a high-fat diet, and, as expected, gain weight and have elevated circulating leptin. We consequently see the predicted elevation in GH secretion, mainly observed via IGF-1. The remainder of the feedback loop described above is not observed, however, in which elevated GH stimulates lipolysis and decreases leptin secretion. This is probably due to the high fat content of the high-fat diet, which prevents a loss of body weight and a fall in leptin levels. Pathophysiologically, when an animal develops leptin resistance, the ability of the hypothalamus to detect circulating leptin is attenuated and GH secretion will be attenuated, as is seen in obesity and which may further contribute to obesity. Assuming this interpretation is correct, no resistance has yet developed in the current study. First, the circulating leptin levels, although elevated in rats fed a high-fat diet, remained within a physiological range. Second, we have seen that GH is still elevated in response to leptin. Last, we observed no down-regulation of leptin receptor mRNA in the hypothalamus. In the chapters of this dissertation that study leptin resistance, we the opposite effects as are demonstrated here: in resistant rats, leptin levels are much higher, GH is attenuated, and hypothalamic leptin receptor is downregulated. 76 In summary, there is a relationship between leptin and GH in the normal animal. We placed young rats on a high-fat diet, which resulted in elevated circulating leptin levels and subsequently elevated endogenous GH and IGF-1 secretion. There are several mechanisms that may explain the occurrence of this phenomenon. Leptin receptors have been found in the pituitary [Cai and Hyde 1998] and leptin may directly regulate GH secretion. Leptin receptors have also been found in the hypothalamus [Mercer et al. 1996] and may regulate the neuropeptides that directly affect GH secretion, GHRH and SRIH. It has also been shown that leptin inhibits neuropeptide Y (NPY) secretion in the ARC [Stephens et al. 1995; Schwartz et al. 1996c]. NPY stimulates SRIH and inhibits GHRH [McCann et al. 1989; Rettori et al. 1990] and therefore inhibits GH, so inhibition of NPY by leptin would prevent the inhibition of GHRH, thereby causing a stimulation of GH. The current study does not demonstrate which, if any or all, of these mechanisms are in effect, but it does show that elevated leptin levels in vivo enhance circulating GH levels, in support of the first hypothesis. CHAPTER 4 LEPTIN TREATMENT INCREASES GROWTH HORMONE SECRETION IN CULTURED GH1 CELLS Introduction Growth hormone (GH) is secreted in an episodic pattern from the somatotropic cells of the anterior pituitary gland. Secretion is under direct hypothalamic control of two neuropeptides: growth hormone releasing hormone (GHRH) and somatotrope release inhibiting hormone (SRIH) [Martin and Millard 1986; Millard 1989]. GHRH stimulates the high amplitude GH secretory episodes. SRIH, on the other hand, is responsible for prolonged intervals during which little GH secretion occurs. SRIH also regulates GHRH secretion [Tannenbaum 1994] and the release of both GHRH and SRIH are, in turn, regulated by many other neuropeptides and neurotransmitters. One of the many roles of GH is regulation of body composition. GH triggers lipolysis [Ho et al. 1996] directly at the level of the adipocyte [Fagin et al. 1980; Vikman et al. 1991]. In obese animals, GH burst amplitudes are reduced [Veldhuis et al. 1991] but administration of recombinant human GH to rats results in a decrease in body weight. The exact mechanism of the attenuation of GH in obesity has yet to be elucidated. Leptin is a hormone produced mainly in adipocytes and which is secreted into the bloodstream in proportion to the amount of fat present. Leptin crosses the blood-brain barrier and informs the hypothalamus of the body's energy stores. The hypothalamus then responds by regulating orexigenic behavior and metabolic rate to maintain body weight homeostasis. 77 78 Both leptin and GH are related to body composition, and it has been shown that circulating leptin and GH are intricately intertwined. A previous study in GH-deficient adults demonstrated a normalization of elevated leptin levels upon GH replacement [Florkowski et al. 1996, Fisker et al. 1997]. This decrease in leptin most likely occurred as a consequence of a decrease in body fat due to GH treatment. Another study showed that leptin treatment induced spontaneous and GHRH-induced GH secretion [Tannenbaum et al. 1998]. Carro et al. [1997] demonstrated that leptin antiserum decreased GH amplitude and nadir in rats suggesting that normal levels of leptin are required for normal GH secretion. The effects of leptin on GH secretion may occur at the hypothalamic level. Leptin receptors colocalize with GHRH neurons [Hakansson et al. 1998] in the arcuate nucleus and are found in the periventricular nucleus [Mercer et al. 1996] where SRIH is made [Kiyama and Emson 1990]. The regulation of either or both of these two hypothalamic GH regulatory factors by leptin may result in indirect control of GH by leptin. In addition, a direct regulation of GH by leptin may occur at the level of the pituitary. It has recently been shown that leptin and leptin receptors are produced in the anterior pituitary [Jin et al. 1999; Jin et al. 2000]. In Chapter 3 we demonstrated that leptin induces GH secretion, but there was no indication of the level of control (hypothalamic or pituitary). The hypothesis of Chapter 4 is that leptin increases GH secretion directly in anterior pituitary cells. To test this hypothesis, a rat pituitary cell line was used. 79 Methods GH1 Cells GH1 cells (American Type Culture Collection, Rockville, MD) are rat pituitary tumor cells that hypersecrete GH. Cells were grown to 70% confluency in F-12K medium (ATCC, Rockville, MD) supplemented with 1% non-essential amino acids, 1% Lglutamine, 1% nystatin (Gibco BRL, Life Technologies, Grand Island, NY), and either 10% horse serum and 2.5% fetal bovine serum (FBS) (Gibco BRL, Life Technologies, Grand Island, NY) or 12.5% charcoal-stripped FBS (Hyclone, Logan, UT). The use of different serum supplementation is described later in the chapter. Cells were incubated at 37ºC with 5% CO2. GH1 cells were used in passes 42-44. Plating Cells and Leptin Concentration In general, cells were plated at 200,000 cells/well in 24-well plates and were allowed time to adhere, usually 2-3 days, before experimentation. On the day prior to each experiment, medium was aspirated from each well and discarded. Control or leptinsupplemented (murine leptin, Amgen, Inc., Thousand Oaks, CA) medium was added to appropriate wells. Leptin was used at a concentration of 100 nM. This concentration of leptin was chosen because 100 nM seems to be in the upper end of the range of physiological leptin levels. Experiments were completed as described below. Five-Day Time -Course Control or leptin-supplemented medium was added to appropriate wells on each plate. Supernatant was collected each day for 5 days and frozen at -35ºC until GH RIA. Cells were collected and analyzed for DNA content for normalization of GH values. 80 Media Experiment Cells were plated using media and supplements as described above, but with differences in serum content. Serum-free media or media supplemented with either 10% horse serum and 2.5% FBS or with 12.5% charcoal-stripped FBS were utilized, as will be described later in the chapter. Supernatant was collected at 8 and 24 hours and frozen at -35ºC until GH RIA. Cells were collected and analyzed for DNA content for normalization of GH values. RT-PCR for Leptin Receptor RNA from GH1 cells was collected for RT-PCR amplification of leptin receptor mRNA as described in Chapter 2. RT-PCR for leptin receptor mRNA was optimized using the sense and antisense primers designed by Zamorano et al. [1997]. The sense and antisense primers were 5’-ATGACGCAGTGTACTGCTG and 5’GTGGCGAGTCAAGTGAACCT, respectively and amplified a 357-base pair fragment of the long form of the leptin receptor. Western Immunoblot for Leptin Receptor Leptin receptor protein expression was observed by Western blotting. Briefly, protein was extracted from hypothalamus, run on a gel, and transferred to a nitrocellulose membrane. The membrane was then incubated with primary antibody to the long form of the leptin receptor at a dilution of 1:500 overnight at 4°C and with secondary antibody for 1 hour at room temperature. Detection reagents were added to the membrane, and the membrane was exposed to radiographic film. The film was developed in a Konica Medical Film Processor (QX-70). 81 GH Radioimmunoassay Cell culture media was measured in duplicate for GH using materials supplied by Dr. A. F. Parlow and the National Hormone and Pituitary Program (NIADDK, Baltimore, MD). Values were expressed in ng/mL in terms of the NIADDK reference preparation rat GHRP-2. The GH RIA has an assay sensitivity of 1 ng/mL and a range of detection of 1 ng/mL to 320 ng/mL. Leptin at concentrations between 1.6 nM and 160 µM do not crossreact with the GH assay. Statistics Student’s t-test was used to compare GH values between time points and between treatment groups. Results Ob-Rb mRNA and Protein Expression The band seen at 357 bp represents mRNA for the long-form of the leptin receptor (Figure 4-1); lanes 3 and 4 are GH1 cells. An additional band, slightly larger, is also seen. This band is most likely a primer-dimer attached to the fragment of interest, which can materialize when the MgCl 2 concentration is too high. This extraneous band was eliminated when the MgCl2 concentration was optimized. For the purposes of this study, we wanted to know whether or not the leptin receptor was present. Therefore the exact concentration of leptin receptor mRNA is not required and no normalization with the housekeeping gene cyclophilin was performed. A Western was completed to determine if the presence of leptin receptor mRNA correlated to leptin receptor protein. The band size (in kDa) representing the long-form of the leptin receptor was not revealed in the 82 Western analysis, however, multiple other bands of smaller sizes, also specific to leptin receptor, were present (data not shown). Five-Day Time -Course Over the 5-day time-course, GH1 cells continued to secrete GH (Figure 4-2) and the GH appeared to be stable in the culture media. Leptin treatment had no effect on GH secretion. Media Experiment In all three serum-supplemented media tested, GH1 cells secreted significantly more GH by 24 hours than at 8 hours. In media with 10% horse serum and 2.5% FBS (Figure 4-3) and in serum-free media (Figure 4-4), leptin had no effect at either time point. In media supplemented with 12.5% charcoal-stripped FBS, leptin significantly increased GH secretion at 8 hours and tended to increase GH at 24 hour (Figure 4-5). 357 bp Figure 4-1: Ob-Rb mRNA on GH1 Cells Lane 1 is the 100 base-pair DNA ladder. Lane 2 is Ob-Rb mRNA in hypothalamus. Lanes 3 and 4 are ObRb mRNA in GH1 cells. 83 Figure 4-2: Five-Day Time-Course Over the 5-day time-course, GH1 cells continually secreted GH (cumulative results over time). Leptin treatment had no effect on GH secretion (t-test, n=6 each). Cells were plated in media supplemented with 10% horse serum and 2.5% FBS. Figure 4-3: Media Supplemented with 10% Horse Serum and 2.5% FBS Significantly more GH was secreted at 24 hours (n=6) than at 8 hours (t-test, p<0.05, n=6). Leptin had no effect at either 8 (n=6) or 24 hours (n=6). 84 Figure 4-4: Serum-Free Media Significantly more GH was secreted at 24 hours (n=6) than at 8 hours (t-test, p<0.05, n=6). Leptin had no effect at either 8 (n=6) or 24 hours (n=6). Figure 4-5: Media Supplemented with 12.5% Charcoal Stripped FBS Significantly more GH was secreted at 24 hours (n=6) than at 8 hours (t-test, * = p<0.05, n=6). Leptin treatment significantly increased GH secretion at 8 hours (t-test, ** = p<0.05) and the effects were nearly significant (p=0.07) at 24 hours (n=6 each). 85 Discussion Leptin has been shown to influence GH secretion indirectly through regulation of hypothalamic neuropeptides [Mercer et al. 1996; Hakansson et al. 1998]. In addition, a direct regulation of GH by leptin may occur at the pituitary level; it has recently been shown that leptin and leptin receptors are produced in the anterior pituitary of humans [Jin et al. 1999] and rodents [Jin et al. 2000]. The current study shows that long-form leptin receptor mRNA is present in GH1 cells. When analyzed for protein expression, the long form of the leptin receptor was not seen, but many other isoforms were. Until recently, it was thought that only the long form of the leptin receptor was biologically active; however, it is now known that the short forms can also transduce signals [Bjorbaek et al. 1997; Murakami et al. 1997; Yamashita et al. 1998]. GH1 cells secrete continuously over time in the absence of a stimulus. In the time-course presented here, GH levels increased consistently over 5 days indicating that GH was stable in the media over time. Leptin treatment had no effect on GH secretion on any of the 5 days. However, these cells were plated in media supplemented with 10% horse serum and 2.5% FBS. After completing these studies, it was discovered that using serum-supplemented media could interfere with the actions of leptin on these cells. To further investigate the effects of serum on leptin treatment, a study was undertaken in which media with different serum contents were utilized. As was seen in the time-course, cells in medium supplemented with 10% horse serum and 2.5% FBS continued to secrete GH over time, but leptin had no effect. There are several potential explanations for this. There may be leptin-binding proteins in the serum that prevent leptin from binding to its cells surface receptors. Conversely, there may be some factor 86 in the serum that antagonizes leptin actions. Additionally, the serum may contain elements that degrade or alter leptin, making it biologically inactive. To eliminate these potentially confounding effects of serum on leptin, serum-free medium was utilized. Again, cells secreted significantly more GH by 24 hours than at 8 hours, but leptin had no effect at either time point. Leptin did, however, tend to decrease GH at 24 hours. This seems contrary to the hypothesis that leptin stimulates GH secretion, however, leptin and GH are substances that may stick to the plastic 24-well plates. In media supplemented with serum, the serum will coat the plates and prevent leptin and GH from sticking. The results of the current study indicated that, in serumfree media, leptin tended to decrease GH secretion. However, it is probable that leptin was bound to the plate and therefore could not produce an effect. In addition, GH may have also adhered to the plate, possibly explaining why there appears to have been less GH secreted in Figure 4-4 than in Figure 4-3. In an attempt to eliminate the uncertainty of the effects of either serumsupplemented or serum-free media on the actions of leptin on GH1 cells, charcoalstripped FBS was utilized. Charcoal stripping reduces the levels of many hormones, growth factors, and steroids. In media supplemented with charcoal-stripped FBS, leptin treatment significantly increased GH secretion at 8 hours, in support of the original hypothesis. Leptin also tended to increase GH secretion at 24 hours, indicating that leptin lost some of its effectiveness over time. With continuous leptin treatment, leptin receptors on GH1 cells may be downregulated. In the future, an 8-hour time-course and a leptin dose-response should be completed. In addition, leptin receptor mRNA concentration could be measured in the normal and leptin-resistant states. 87 The GH data reported was normalized to DNA content. It was found that leptin treatment of GH1 cells in charcoal-stripped media decreased DNA content at both 8 and 24 hours, indicating that cell proliferation was inhibited. These results agree with those in which leptin treatment decreased proliferation in GH3 cells [Jin et al. 2000]. The GH3 cell line was initiated from the same primary culture from which the GH1 cells were initiated, two passages later. When we analyzed raw GH data, leptin had no effect (data not shown) but when we analyzed GH data normalized to DNA, leptin stimulated GH secretion. Taken together, these results suggest that leptin inhibits cell proliferation but stimulates GH secretion from the cells that are present. GH1 cells are tumor cells, so any extrapolation to in vivo physiology is uncertain; however, perhaps these results indicate that leptin stimulates GH while decreasing cell proliferation as a self-limiting process. This should be studied in the future in primary pituitary cells. In vivo, serum is not charcoal-stripped, nor is there a serum-free option, so it may be argued that the methods utilized in this chapter are not indicative of what happens in whole animal physiology. In animals, serum is present that contains leptin-binding proteins. However, it is possible that these proteins have a lower affinity for leptin than leptin receptors, so when bound leptin reaches the desired destination, the binding proteins would release leptin making it available to act at its receptor. In summary, the studies presented here indicate that GH1 cells express leptin receptor mRNA and that leptin stimulates GH secretion from these cells. These results support the first hypothesis of this dissertation. CHAPTER 5 LEPTIN RESISTANCE IS ASSOCIATED WITH HYPOTHALAMIC RECEPTOR mRNA AND PROTEIN DOWNREGULATION Introduction The ob gene expresses the "obese protein," leptin, which is secreted from adipocytes and circulates in the plasma. Leptin has been implicated as one of the signals that is sensed by the hypothalamus to regulate feeding and energy expenditure thereby maintaining body weight homeostasis [Zhang et al. 1994; Campfield et al. 1995; Halaas et al. 1995; Pelleymounter et al. 1995; Halaas et al. 1997]. Leptin is expressed primarily in adipocytes, including those of epididymal, parametrial, abdominal, perirenal, and inguinal fat pad origin [Maffei et al. 1995a; McGregor et al. 1996] and, to a much lesser extent in brown adipose tissue [Maffei et al. 1995a]. Moreover, leptin is secreted in proportion to adipose mass [Considine et al. 1996b; Maffei et al. 1995b]. Leptin receptors are located in many peripheral tissues [Cioffi et al. 1996; Lee et al. 1996; Tanizawa et al. 1997] and the central nervous system (CNS). In the CNS, leptin receptors are especially abundant in areas of the hypothalamus that are implicated in body weight regulation: arcuate, ventromedial, paraventricular, and ventral premammillary nuclei [Mercer et al. 1996]. Many splice variants of the receptor have been found. Most isoforms have short intracellular domains, some of which are thought to regulate transport of leptin into the brain. Other roles for these isoforms have yet to be elucidated. Another type of splice variant has a longer intracellular domain. This is the variant through which leptin exerts 88 89 a biological effect. Recently, a soluble isoform of the leptin receptor has been characterized which has been proposed to regulate leptin bioactivity [Liu et al. 1997]. A mutation of any of these isoforms could result in resistance to leptin, as is seen in the obese fa/fa rat and db/db mouse. Mice with mutations of the ob gene have reduced levels of circulating leptin and exhibit hyperphagia and obesity. When these animals are given exogenous leptin, their body weight is significantly reduced. Unfortunately, these findings are not easily extrapolated to human obesity. Many obese humans exhibit dramatically elevated levels of circulating leptin [Zhang et al. 1994; Maffei et al. 1995b] that can result in an inability of the hypothalamus to detect leptin. When this occurs, the result is increased feeding, added fat mass, and further elevated levels of circulating leptin. Failure of the hypothalamus to detect leptin, or leptin resistance, may occur for one of several reasons. First, leptin may fail to cross the bloodbrain barrier. Secondly, the hypothalamic receptors may be mutated or downregulated. Alternatively, downstream signaling may be inhibited. Although investigators are not all of the same opinion, most investigators believe in the development of a leptin-resistant state with the progression toward obesity in both humans [Caro et al. 1996a] and animals [Frederich et al. 1995a; Maffei et al. 1995b; Van Heek et al. 1997]. The current study demonstrates that resistance to the anorectic and GH-regulating effects of leptin develops in conjunction with downregulation of hypothalamic receptor in male Long-Evans rats. Methods Animals Thirty-six male Long-Evans rats (150-174 g, 42-45 days, Harlan Sprague-Dawley) were obtained from Harlan Sprague-Dawley and housed individually under standard lighting 90 and temperature conditions. The rats were fed normal chow, which, along with water, was available ad libitum. The animals were randomly divided into 3 leptin-treated groups. Body weight and food intake were measured daily for the duration of the study. Groups Three groups of animals (12 per group) were implanted subcutaneously with Alzet osmotic pumps (Model 2ML4, Alza Scientific Products, Palo Alto, CA) that deliver their contents at a rate of 2.5 µL/hr for 28 days. The contents of the pumps consisted of sterile PBS (control), leptin at a dose of 0.1 mg/kg/day (low dose), or leptin at a dose of 0.5 mg/kg/day (high dose). The lower dose was chosen to mimic physiological leptin levels in obese rats (10-25 ng/mL) while the higher dose was chosen to test for resistance in the event that it was not seen at the lower dose. The murine leptin used in this study was provided by Amgen, Inc. (Thousand Oaks, CA). Serum Measurements Blood samples were collected by cardiac puncture on the day of implantation (day 1) and on day 15; trunk blood was collected at sacrifice (day 29) for determination of leptin, insulin-like growth factor-1 (IGF-1), insulin, glucose, and triglycerides. All samples were collected in the morning between 8 AM and 12 PM. Leptin was measured with a rat leptin radioimmunoassay (RIA) kit (Linco Research, Inc., St. Charles, MO). This kit measures both rat and mouse leptin with an assay sensitivity of 0.5 ng/mL and a range of detection from 0.5 ng/mL to 50 ng/mL. IGF-1 was extracted from blood by the acid/ethanol procedure and measured by RIA as previously described [Grant et al. 1986]. The IGF-1 RIA has an assay sensitivity of 0.1 ng/mL and a range of detection from 0.1 ng/mL to 20 ng/mL. Insulin was measured with a rat insulin RIA kit (Linco Research, Inc., St. Charles, MO) with an assay sensitivity of 0.1 ng/mL and a range of detection 91 from 0.1 ng/mL to 10 ng/mL. Glucose was measured using a YSI 1500 Sidekick Glucose Analyzer (Yellow Springs Instrument Company, Inc., Yellow Springs, OH). Triglycerides were measured using a kit from Sigma (Sigma Diagnostics, St. Louis, MO). Leptin Challenge Test Pilot Study Dose and route of leptin administration were determined in a pilot study prior to the actual resistance study. Naïve rats were food deprived for 24 hours then given an intraperitoneal (IP) or subcutaneous (sc) leptin or PBS injection, utilizing different doses of leptin. In two studies, leptin was given IP at either 1.5 or 2.8 mg/kg. In another study, 30 mg/kg leptin was given sc. Food was returned after each injection, and food intake was measured at 24 hours. Leptin Challenge Test A test of leptin resistance was performed on day 21. The animals were food deprived for 24 hours prior to the test. The leptin challenge test consisted of a sc bolus of murine leptin (30 mg/kg) which was given to half of the animals in each treatment group (leptin challenge). The other half of the animals in each group received a bolus of PBS for control. Food (Purina rat chow #5001) was returned to the animals one hour postinjection, allowing the animals time to recover following the injection. Food intake was then measured at 4 hours and again at 24 hours. RT-PCR for Leptin Receptor RT-PCR for leptin receptor mRNA was optimized using the sense and antisense primers from Zamorano et al. [1997]. The sense and antisense primers were 5’ATGACGCAGTGTACTGCTG and 5’-GTGGCGAGTCAAGTGAACCT, respectively and amplified a 357-base pair fragment of the long form of the leptin receptor. Optimal conditions were identified as 1 mM MgCl 2, 64°C annealing temperature, and 30 cycles 92 following an initial denaturation phase and followed by a final elongation phase. The identity of RT-PCR product was confirmed using the internal probe 5’TGCAGCTGAGGTATCACAGG in Southern blot analysis (ECL, Amersham Pharmacia Biotech, Piscataway, NJ). The amount of leptin receptor mRNA was normalized with the housekeeping gene cyclophilin. The cyclophilin sense and antisense primers used were 5'-GGGAAGGTGAAAGAAGGCAT and 5'-GAGAGCAGAGATTACAGGGT, respectively and amplified a 210-base pair fragment [Zamorano et al. 1997]. The same conditions were used for cyclophilin as were used for the leptin receptor. Western Immunoblot for Leptin Receptor Leptin receptor protein expression was measured by Western blotting. Briefly, protein was extracted from hypothalamus, run on a gel, and transferred to a nitrocellulose membrane. The membrane was then incubated with primary antibody at a dilution of 1:500 overnight at 4°C and with secondary antibody for 1 hour at room temperature. Detection reagents were added to the membrane, and the membrane was exposed to radiographic film. The film was developed and analyzed with Molecular Analysis Software (Hercules, CA). The results were normalized according to the amount of protein loaded, and the results are expressed as a ratio of sample:intra-gel control. Statistics Body weight, food intake, IGF-1 levels, and leptin values were analyzed by two-way repeated measures ANOVA followed by Student-Newman-Keuls Multiple Comparisons. Comparisons between the leptin challenge and control groups were by Student's t-test for each pilot study. Two-way ANOVA and Duncan's Multiple Comparisons were used to analyze food intake in response to the leptin challenge. One-way ANOVA followed by 93 Tukey’s or Dunn's Multiple Comparisons was used to analyze insulin, glucose, and triglyceride, and leptin receptor mRNA and protein expression data. Results Leptin Leptin levels were significantly elevated in the low-dose implant group after 4 weeks of treatment as well as in the high-dose implant group after 2 and 4 weeks of treatment compared to control (Figure 5-1). At time of implantation, there were no differences in serum leptin levels among the groups. Within the high-dose group, leptin levels were significantly elevated at weeks 2 and 4 compared to week 0. Body Weight Rats in the control group weighed significantly more than rats in the high-dose group at day 7 and from day 9 through the remainder of the study (Figure 5-2). Body weight between the two leptin-treated groups was significantly different at days 12, 15-19, and 21-29. The differences in body weight between the low-dose group and the controls never reached statistical significance although there was a trend for the low-dose rats to weigh less than the control rats. All 3 groups experienced a drop in body weight following 24-hour periods of food deprivation, indicated by discontinuations of graph lines. Food Intake Rats in the high-dose group consumed significantly less rodent chow than controls on days 4-9, 16, and 23 (Figure 5-3). Rats in the low-dose group also ate significantly less than controls on days 4-6 and 23. The difference in food intake in the two leptin-treated 94 groups was significantly different at days 4-9. Periods of food deprivation are indicated by discontinuations of graph lines. Leptin Challenge Test Pilot Studies Neither the 1.5 mg/kg nor the 2.8 mg/kg dose of leptin administered IP was effective at reducing food intake at 24 hours. Leptin given sc at a dose of 30 mg/kg reduced 24-hour food intake significantly compared to control (Figure 5-4). This dose and route of leptin administration (leptin challenge) was used in the leptin challenge test. Leptin Challenge Test At 24 hours, the rats in the control implant group ate significantly less when challenged with leptin compared to their respective controls whereas the leptin challenge had no effect in rats treated with either dose of leptin (Figure 5-5). In addition, there were no differences in food intake among the treatment groups due to the leptin challenge, but food intake of the low- and high-dose leptin groups were significantly lower than controls following PBS control injection. Leptin Receptor mRNA in Hypothalamus In both leptin-treated groups, leptin receptor mRNA was significantly reduced when compared to control (Figure 5-6). Leptin receptor mRNA was normalized to the housekeeping gene cyclophilin. Leptin Receptor Protein Expression in Hypothalamus The high-dose leptin-treated group had significantly reduced leptin receptor protein expression compared to control (Figure 5-7). The low-dose leptin group also tended to have less protein expression compared to control, but the results were not significant. 95 IGF-1 Values At week 2, the high-dose leptin group had significantly reduced IGF-1 values compared to week 0 (Figure 5-8). At week 4, both the control and low-dose leptin groups had significantly elevated IGF-1 values compared to week 0. Also at week 4, IGF-1 was significantly lower in the high-dose leptin group than in either of the other two groups. Hormonal and Metabolic Measures Measures of hormonal and metabolic state including insulin, glucose, and triglycerides are reported in Table 5-1. At both weeks 2 and 4, triglycerides were significantly attenuated in the high-dose leptin group compared to the other two groups. At week 4, glucose was significantly reduced in the high-dose group compared to the other two groups. Also at week 4, insulin was significantly lower in both the low- and high-dose groups compared to control. Figure 5-1: Leptin Levels Indicate Proper Pump Activity Serum leptin levels were significantly elevated (* = p<0.05 vs. control) in the low dose treatment group (n=12) compared to control (n=12) and in the high-dose treatment group (n=12) at weeks 2 and 4 compared to control (2-way repeated measures ANOVA). Serum leptin levels were significantly higher (# = p<0.05 vs. week 0) in the high-dose treatment group at weeks 2 and 4 compared to week 0. 96 Figure 5-2: Body Weight is Dose-Dependently Decreased by Leptin Treatment There were no significant differences in body weight between the control (n=12) and low dose treatment groups (n=12) although the low dose group tended to weigh less for the duration of the study (2-way repeated measures ANOVA). Body weight in the high dose treatment group (n=12) was significantly decreased (p<0.05) at days 12, 15-19, and 21-29 compared to the low dose group and at days 7 and 9-29 compared to control (2-way repeated measures ANOVA). Discontinuations of graph lines follow 24-hour periods of food deprivation resulting in erroneous decline in body weight. Figure 5-3: Food Intake is Initially Decreased by Leptin; Ultimately Resistance Develops Food intake was significantly inhibited (p<0.05) in the low dose treatment group (n=12) compared to control (n=12) at days 4-6 and 23 (2-way repeated measures ANOVA) and in the high dose treatment group (n=12) compared to control at days 4-9, 16, and 23. Food intake was significantly different (p<0.05) between the two treatment groups at days 4-9 (2-way repeated measures ANOVA). Discontinuations of graph lines represent 24-hour periods of food deprivation. 97 Figure 5-4: Leptin Challenge Test Pilot Study In the pilot study which used a leptin challenge of 30 mg/kg administered subcutaneously (sc), leptin significantly reduced food intake (p<0.05, Student's t-test) compared to control. Neither the 1.5 mg/kg nor the 2.8 mg/kg intraperitoneal (IP) leptin challenge was effective at reducing food intake. Each group, n=6. Figure 5-5: Leptin Challenge Test At 24 hours, food intake was significantly inhibited (p<0.05) by leptin challenge (n=6) compared to respective controls (n=6) within the treatment control group (2-way ANOVA). There were no differences in food intake as the result of leptin challenge (n=6) in either leptin-treated group compared to respective controls (n=6). Food intake in the control treatment group was significantly higher than in their low- and high-dose leptin counterparts following the PBS control injection. 98 Figure 5-6: Leptin Receptor mRNA in Hypothalamus In both the low-dose (n=12) and high-dose (n=10) leptin groups, leptin receptor mRNA was significantly reduced in hypothalamus (p<0.05, 1-way ANOVA) compared to control (n=12). Leptin receptor mRNA was normalized to the housekeeping gene cyclophilin. Figure 5-7: Leptin Receptor Protein Expression in Hypothalamus The high-dose leptin-treated group (n=9) had significantly reduced leptin receptor protein expression compared to control (p<0.05, 1-way ANOVA, n=12). The low-dose leptin group (n=9) also tended to have less protein expression compared to control, but the results were not significant. 99 Figure 5-8: IGF-1 Values At week 2, the high-dose leptin group had significantly reduced IGF-1 values compared to week 0 (p<0.05, 2-way repeated measures ANOVA). At week 4, both the control and low-dose leptin groups had significantly elevated IGF-1 values compared to week 0. Also at week 4, IGF-1 was significantly lower in the high-dose leptin group than in either of the other two groups. N=8-12. Table 5-1: Hormonal and Metabolic Measures Week 0 Insulin Glucose (mg/dL) (ng/mL) Control N=10-12 1.96 ± 0.24 177 ± 4.93 Low Leptin N=8-12 1.73 ± 0.24 176 ± 5.05 High Leptin N=7-12 2.28 ± 0.24 164 ± 4.16 Week 2 Control Low Leptin High Leptin Glucose (mg/dL) N=9-10 N=3-8 N=3-8 Insulin (ng/mL) 1.04 ± 0.18 0.41 ± 0.11 0.81 ± 0.13 Glucose (mg/dL) N=12 N=12 N=8-12 Insulin (ng/mL) 1.72 ± 0.16 1.17 ± 0.12 * 0.77 ± 0.15 * Week 4 Control Low Leptin High Leptin 168 ± 3.36 162 ± 3.34 162 ± 1.53 158 ± 3.29 156 ± 2.42 144 ± 2.75 ** Triglycerides (mg/dL) 103.20 ± 8.10 110.09 ± 9.08 107.31 ± 8.25 Triglycerides (mg/dL) 81.37 ± 5.88 72.99 ± 8.22 47.66 ± 4.59 ** Triglycerides (mg/dL) 122.16 ± 7.00 98.77 ± 9.89 57.73 ± 9.04 ** Values are given as mean ± SEM. * = p<0.05 vs. control. ** = p<0.05 vs. control and low leptin. 1-way ANOVA. 100 Discussion At time of osmotic pump implantation (week 0), there were no differences in serum leptin levels among the different treatment groups. At week 2, the high-dose group had significantly elevated leptin levels compared to control and to the low-dose group. At week 4, both the high-dose and the low-dose groups had significantly elevated leptin levels compared to control. In addition, serum leptin levels were significantly elevated in the high-dose leptin-treated group at weeks 2 and 4 compared to week 0. Taken together, these results indicate that the osmotic pumps were effective at delivering their contents and the murine leptin was stable. Additionally, leptin has been shown to be structurally stable after 4 weeks at 37°C (MaryAnn Pelleymounter, Neurocrine Biosciences, Inc., personal communication). It was previously shown that, within 24 hours of leptin withdrawal, body weight normalizes immediately (MaryAnn Pelleymounter, Neurocrine Biosciences, Inc., personal communication). A normalization of body weight was not observed in the current study indicating that leptin continued to be effective over time; in addition to its stability, leptin maintained its biological activity. With continuous infusion of drug it would be expected that, once established, steady state serum levels would be maintained over time. On the contrary, leptin levels in these rats continued to increase throughout the study period, indicating that steady state was never achieved. One potential mechanism by which this occurred could have been a saturation of clearance or a downregulation of "clearance" receptors (Ob-Ra on kidney); however, it was previously shown that, even when pharmacological doses of leptin are administered, elimination pathways are not saturated [Cumin et al. 1996; Hill et al. 1998]. It is unknown if the doses used in this dissertation exceeded the pharmacological 101 doses used in saturation or elimination studies. The more likely explanation of lack of steady state leptin levels is the result a pseudo-two-compartment model of drug infusion. Upon implantation, cysts rapidly form around the osmotic pumps. As expected, some of the leptin being released from the pump gets into the bloodstream. In addition, some of the leptin may also be sequestered by the cyst. Over time, as the cyst gets saturated with leptin, leptin would be released from the cyst as well as from the pump. This would result in higher serum leptin levels over time, as is seen in the current study. It is generally believed that with leptin treatment food intake is attenuated. This was seen in the first two weeks of our study in a dose-dependent manner: the animals treated with high-dose leptin ate significantly less than those treated with low-dose leptin, which in turn ate less than controls. During the final two weeks of the study, there were virtually no differences in food intake among the groups indicating that the rats in each treatment group became resistant to the long-term appetite regulating effects of leptin. This loss of effect of leptin treatment on food intake over time was shown previously by us (unpublished observations) and others [Pelleymounter et al. 1995]. The mechanism by which the anorectic effects of leptin were lost is likely the downregulation of hypothalamic leptin receptor mRNA and protein that occurred as a pharmacological response to the higher levels of circulating leptin. It appeared that the leptin-treated rats began to develop resistance to the anorectic effects of leptin approximately half way through the study, so we performed a leptin challenge test at week 3. The leptin challenge test was a one-time sc bolus of a dose of leptin high enough to attenuate 24-hour food intake in control rats. Within each treatment group, half of the animals received the leptin challenge and the other half 102 served as controls and received an injection of PBS. As expected within the control treatment group, the leptin challenge acted to decrease food intake at 24 hours. Alternatively, the rats within the leptin treatment groups did not respond to the leptin challenge; there were no significant differences in food intake in either treatment group at either time point following the leptin challenge compared to their respective controls. These results, together with the food intake results, indicate that continuous exogenous leptin treatment results in the development of resistance to the appetite regulating effects of leptin by the 2nd or 3rd week of treatment. Additionally, in the 4-hour leptin challenge test in the low-dose leptin group, there was a statistically nonsignificant increase in food intake in response to the leptin challenge. By 24-hours, however, there was no detectable effect of leptin on food intake in the chronically treated groups. An important observation with this test must be discussed. The control rats in the chronic PBS treatment group ate significantly more than the control rats in either chronic leptin treatment group. These results suggest that, although resistance to the appetiteregulating effects of leptin was developing in the animals treated chronically, the longterm leptin treatment resulted in some sort of ceiling effect is seen in which the rats simply cannot eat as much as untreated rats. In addition, the leptin-treated animals were eating the same amount as the untreated animals at this time point. Leptin treatment decreases or attenuates increases in body weight. At the beginning of the current study, leptin treatment dose-dependently decreased body weight, which corresponded with the inhibitory effects on food intake. After the second week, leptin did not cause further decrease in body weight and the rate of growth was similar in all three groups indicating that “catch-up” growth was prevented. Unlike the food intake 103 effects of leptin, the animals did not become resistant to the body weight regulating effects of leptin. Our body weight and food intake results correspond to those from a study in which mice, after receiving daily leptin injections, experienced a normalization of the initially reduced food intake [Pelleymounter et al. 1995]. In that study, the reduction in body weight was never normalized. In this chapter, the body weight never normalized. IGF-1 has a relatively long half-life and is secreted more continuously than GH and therefore is a useful indicator of GH secretory status [Blum et al. 1990]. In the control and low-dose leptin groups, IGF-1 levels were significantly elevated by week 4. This may be explained in the controls due to normal growth. The results of the IGF-1 seen in the low-dose leptin group support the first hypothesis of the dissertation. It may be expected, however, that since the low-dose group had much higher leptin levels than the control group, the low-dose group should also have significantly elevated IGF-1 values compared to the control group. This was not seen, perhaps indicating that resistance to the GH-stimulating effects of leptin were beginning to be lost. In support of this suggestion, hypothalamic leptin receptor protein expression was apparently downregulated, although the effects were not significant. In the high-dose leptin group, IGF-1 values declined over time and were lower than IGF-1 in either of the other two groups at week 4. The results of Figure 5-8 indicate that, at this length of time of pump implantation, the lower dose of leptin was not sufficient and higher dose of leptin was sufficient to cause resistance to the GH-stimulating effects of leptin. There are different mechanisms by which food intake and body weight are regulated by leptin, including central alterations in metabolic rate and energy expenditure 104 as well as direct effects on fat cells. Neither metabolic rate/energy expenditure nor fat content were measured in this study. We did, however, measure other indices of metabolic action and found decreases in serum insulin, glucose, and triglyceride levels in the high-dose leptin group as well as a decrease in insulin in the low-dose group by week 4. It should be pointed out the basal glucose in the control rats is elevated, probably due to the stressful sampling technique. The results of Table 5-1 indicate that leptin retained its ability to affect metabolic parameters even though its ability to alter food intake was lost. The results of the food intake, leptin challenge, IGF-1, and metabolic studies indicate that the behavioral and metabolic aspects of leptin resistance occur by different mechanisms and at different levels of circulating leptin. One type of leptin resistance occurs as the result of a mutation of the leptin receptor. There are various types of leptin receptor mutations that result in obesity in rodents [Flier and Elmquist 1997] as well as in humans [Clement et al. 1998]. Leptin resistance can also develop over time as opposed to being the result of a mutation. In a recent study in obese mice with peripheral resistance to leptin, it was shown that central leptin administration had the desired effect of reduction of food intake and body weight [Van Heek et al. 1997]. These results demonstrate that leptin resistance may occur if leptin progressively loses its ability to cross the blood-brain barrier. Transport of leptin across the blood-brain barrier is unidirectional from blood to brain and occurs by a saturable process [Banks et al. 1996; Caro et al. 1996a] via short-form leptin receptors. Perhaps in response to the slowly developing hyperleptinemia that occurs with increasing body weight, there is a down-regulation of these leptin transporters [Caro et al. 1996a]. It must be noted, however, that several of the hypothalamic nuclei where leptin exerts many 105 of its biological effects are circumventricular organs, located near fenestrations of the blood-brain barrier. Other forms of leptin resistance can occur in a pharmacokinetic manner, for example if there is a problem with distribution, delivery, metabolism, and/or elimination. Leptin resistance can also occur in a pharmacodynamic manner, with receptor downregulation or disruption of signal transduction. More than likely, there are multiple potential mechanims for the development of leptin resistance. It has been shown there are varying degrees of leptin resistance among different strains of obese mice [Halaas et al. 1995] and there are likely to be variations of leptin resistance in obese humans as well. There are multiple complex CNS pathways through which leptin produces bioactivity [Elmquist et al. 1998], and a problem with any one of them can result in a perturbation of body weight homeostasis. An important part of the mechanism(s) by which leptin resistance occurred in our study incorporated downregulation of hypothalamic leptin receptor mRNA and protein expression. It is a well-known pharmacological phenomenon that elevated levels of agonist, in this case circulating leptin, result in downregulation of receptor to prevent excessive activity. When this occurrence is chronic, there develops a state of resistance to the agonist. This response is analogous to insulin resistance seen in hyperinsulinemia. There is a strong historical precedence for agonist-induced receptor downregulation and our findings on leptin receptor downregulation support this model of resistance. This is an important and heuristic finding. In conclusion, we have demonstrated the development of resistance to the anorectic and GH-regulating effects of leptin in conjunction with downregulation of 106 hypothalamic leptin receptor mRNA and protein expression. These results support the second hypothesis of this dissertation. CHAPTER 6 LEPTIN'S EFFECTS ON FOOD INTAKE AND BODY WEIGHT ARE DIFFERENTIALLY ATTENUATED IN RATS FED A LOW-FAT OR HIGH-FAT DIET Introduction Leptin is a hormone secreted primarily by adipose tissue [Maffei et al. 1995a; McGregor et al. 1996]. It travels in the bloodstream, actively crosses the blood-brain barrier, and binds to its receptors in the hypothalamus to inform the brain of the body’s energy stores. Leptin levels increase in parallel with body fat [Considine et al. 1996b; Maffei et al. 1995b]. When the brain senses this elevation of leptin levels, the response is decreased feeding and heightened metabolic rate [Zhang et al. 1994; Campfield et al. 1995; Halaas et al. 1995; Pelleymounter et al. 1995; Halaas et al. 1997]. Conversely, the absence of leptin that is seen in the ob/ob mouse or defective leptin signaling that is seen in the db/db mouse and the fa/fa rat results in obesity due to hyperphagia and a low metabolic rate. Exogenous leptin administered to leptin-deficient obese animals results in a lowered body weight due to the restoration of normal feeding patterns and metabolism [Campfield et al. 1995; Halaas et al. 1995; Pelleymounter et al. 1995]. In addition, the reduction in body weight is observed when leptin is administered to leptin-producing animals of normal body weight. Leptin’s behavioral effects are attenuated in animals fed a highly palatable diet [Campfield et al. 1995; Frederich et al. 1995a; Masuzaki et al. 1995; Van Heek et al. 1997; Widdowson et al. 1997] even though leptin is increased. It would be expected that 107 108 the elevated leptin levels would cause a decrease in food intake and body weight [Campfield et al. 1995], but conversely, food intake and body weight were elevated in these studies [Frederich et al. 1995a; Masuzaki et al. 1995; Van Heek et al. 1997; Widdowson et al. 1997]. Studies completed to date examined elevated leptin levels that originated from endogenous sources and focused on the effects of high-fat diets. The current study examines the effects of exogenously elevated serum leptin and was undertaken to test the hypothesis that induced hyperleptinemia, due to exogenous leptin administration, also fails to be effective in rats fed a high-fat, high-calorie diet. Furthermore, the study was designed to ascertain if the same would be observed in rats fed a low-fat, high-calorie diet. The same diets were used in this study as were used in Chapter 3, but the animals were fed for a longer period of time before experimentation to allow the development of resistance. In addition, this study utilizes continuous delivery of leptin via osmotic minipumps, as was used in Chapter 5. In this chapter, leptin was given for four weeks at a dose between the two doses used previously. Methods Animals and Diets Twenty-two male Long-Evans rats (90-95 days, 350-450 g) were obtained from HarlanSprague Dawley and housed individually under standard temperature and lighting conditions. The animals were randomly divided into three groups (n=7 or 8), each receiving varied diets ad libitum: normal rat chow (5% fat, 3.0 kcal/g, Purina Mills, Inc., Richmond, VA), high-fat, high-calorie diet (23% fat, 3.7 kcal/g, PJ Noyes Company, Inc., Lancaster, NH), or low-fat, high-calorie diet (2% fat, 3.7 kcal/g, PJ Noyes Company, Inc., Lancaster, NH). It should be noted that the low-fat diet had a calorie 109 content equivalent to that of the high-fat diet, both of which were higher in calories than the control diet. Water was available to all groups ad libitum. Body weight and food intake were measured daily for the duration of the study. Rats were sacrificed by decapitation and the weights of brains, pituitaries, hearts, testes, kidneys, livers, and adrenals were measured. Hypothalami were snap frozen in liquid nitrogen and stored at -90°C until further analysis. Osmotic Pumps, Blood Sampling, and Body Temperature After 50 days on the various diets, half the rats in each group were implanted subcutaneously with leptin-filled Alzet osmotic mini-pumps (Alza Scientific Products, Palo Alto, CA) for the continuous delivery of leptin (0.25 mg/kg/day; 5 µg/hour) over a 14-day period; the other half were implanted with pumps delivering vehicle (control). Murine leptin was provided by Amgen, Inc. (Thousand Oaks, CA). Blood samples were collected from the tail vein prior to pump implantation (week 0) and at week 1 and trunk blood was collected at week 2 for measurement of leptin and IGF-1. All samples were collected in the morning between 8 AM and 12 PM. Blood was centrifuged and serum was stored at -20°C until analysis. Body temperature was measured at weeks 0, 1, and 2 with an anal thermistor (Cole Parmer, Vernon Hills, IL). Leptin and IGF-1 Radioimmunoassays Serum leptin was measured with a rat leptin RIA kit (Linco Research, Inc., St. Charles, MO). This kit measures both rat and mouse leptin with an assay sensitivity of 0.5 ng/mL and a range of detection from 0.5 ng/mL to 50 ng/mL. IGF-1 was extracted from blood by the acid/ethanol procedure and measured by RIA as previously described [Grant et al. 110 1986]. The IGF-1 RIA has an assay sensitivity of 0.1 ng/mL and a range of detection of 0.1 ng/mL to 20 ng/mL. Statistics Body weight, food intake, and leptin values were analyzed by two-way repeated measures ANOVA followed by Student-Newman-Keuls Multiple Comparisons. Twoway ANOVA was used to analyze temperature each week and organ weights, and IGF-1 values followed by Duncan's Multiple Comparisons. Results Effects of Diet on Food Intake Figure 6-1 shows absolute food intake for the duration of feeding of the three different diets. Since there are differences in kcal and percent fat in each diet, food intake is reported in grams of food (panel A), kcal (panel B), and grams of fat (panel C) ingested. The normal chow rats ingested more grams of food than the other two groups for a majority of the study, but there were no differences in the intake of kcal among the three groups. As expected, the high-fat fed rats ingested the highest amount of fat while the low-fat rats ingested the lowest. Leptin Basal serum leptin levels were not different among the PBS-implanted groups (Figure 62). As expected, leptin levels did not change significantly over the 2-week implantation time in rats of any diet group implanted with control pumps. In the rats implanted with leptin-filled osmotic pumps, serum leptin levels increased significantly in each of the groups over the two-week period. 111 Effects of Diet and Leptin on Food Intake As expected, leptin treatment decreased food intake (measured in grams of food) in rats fed normal chow for most of the study (Figure 6-3 panel a). Unexpectedly, on days 7, 12, and 13, leptin treated actually increased food intake in rats fed the high-fat diet (Figure 63 panel b). There was no effect of leptin on food intake in rats fed the low-fat diet (Figure 6-3 panel c). Food intake was also shown as grams of food normalized to 100 g body weight (Figure 6-4). Leptin treatment decreased food intake in rats fed normal chow on days 2-6 and 10 (panel A); however, food intake in the leptin-treated group equaled that in the PBS group by the end of the 2 week implantation period. Leptin had virtually no effect in rats fed the high-fat (panel B) and had little effect on rats fed the low-fat diet (panel C). Effects of Diet and Leptin on Body Weight The change in body weight was calculated by subtracting the daily body weight from body weight on the day of pump implantation (prior to surgery) for individual animals, then an average was taken. Change in body weight was used instead of raw body weight data due to the differences in starting body weights of the animals within the treatment groups. In rats fed normal chow, the expected leptin-induced decrease in body weight was observed beginning on day 3 of leptin administration and continuing for the duration of the study (Figure 6-5). Leptin treatment in rats fed a high-fat diet, however, attenuated body weight only on days 10 and 11. In animals receiving the low-fat diet, body weight was significantly lower in controls starting on day 4 of leptin treatment and continuing through day 14. 112 Effects of Diet and Leptin on IGF-1 Values In rats fed normal chow, IGF-1 was attenuated by leptin treatment (Figure 6-6) whereas leptin had no effect on IGF-1 in rats fed the low-fat diet. In rats fed the high-fat diet, IGF-1 was decreased in both the leptin-treated and PBS-treated groups. Effects of Diet and Leptin on Organ Weights Weights of organs were normalized per 100 grams body weight for each individual rat. There was no difference in weights of brains, pituitaries, hearts, testes, kidneys, livers, or adrenals in any groups of animals at any time point (Table 6-1). Effects of Diet and Leptin on Body Temperature There were no significant differences in body temperature in any groups of rats at any time point (Table 6-2). 113 Figure 6-1: Absolute Food Intake Reported in Various Diet Parameters Food intake reported as grams of food in rats fed a high-fat diet, a low-fat diet, and normal rat chow (panel A). Food intake reported as kilocalories in rats fed a high-fat diet, a low-fat diet, and normal rat chow (panel B). Food intake reported as grams of fat in rats fed a high-fat diet, a low-fat diet, and normal rat chow (panel C). 114 Figure 6-2: Leptin Levels Indicate Proper Pump Activity In rats fed normal chow serum leptin levels were significantly elevated (p<0.05) at week 2 of leptin treatment (n=4) (2-way repeated measures ANOVA, panel A). In rats fed the high-fat diet serum leptin levels were significantly elevated (p<0.05) at weeks 1 and 2 in response to leptin treatment (n=4) (2-way repeated measures ANOVA, panel B). In rats fed the low-fat diet serum leptin levels were significantly elevated (p<0.05) at week 2 of leptin treatment (n=4) (2-way repeated measures ANOVA, panel C). 115 Figure 6-3: The Effects of Leptin on Food Intake in Diets of Varying Calorie and Fat Contents In rats fed normal chow food intake was significantly reduced (p<0.05, 2-way repeated measures ANOVA) in leptin-treated rats (n=4) on days 2-11 and 14 compared to control (n=3, panel A). In rats fed t he high-fat diet food intake was significantly elevated (p<0.05, 2-way repeated measures ANOVA) in leptin-treated (n=4) rats on days 7, 12, and 13 compared to control (n=4, panel B). In rats fed the low-fat diet there was no effect of leptin treatment (n=4) compared to control (p<0.05, 2-way repeated measures ANOVA, panel C). 116 Figure 6-4: The Effects of Leptin on Food Intake - Normalized to 100 grams Body Weight In rats fed normal chow food intake was significantly reduced (p<0.05, 2-way repeated measures ANOVA) in leptin-treated rats (n=4) on days 2-6 and 10 compared to control (n=3, panel A). In rats fed the high-fat diet food intake was significantly reduced (p<0.05, 2-way repeated measures ANOVA) in leptin-treated (n=4) rats on day 4 compared to control (n=4, panel B). In rats fed the low-fat diet food intake was significantly reduced (p<0.05, 2-way repeated measures ANOVA) in leptin-treated (n=4) rats on days 2, 3, and 11 compared to control (n=3, panel C). 117 Figure 6-5: The Effects of Leptin on Body Weight in Diets of Varying Calorie and Fat Contents In rats fed normal chow PBS-treated rats (n=3) gained weight during the 14-day period; leptin-treated rats (n=4) did not. The results were significantly different (p<0.05) beginning at day 3 of treatment of continuing for the duration of the study (2-way repeated measures ANOVA, panel A). In rats fed the highfat diet both leptin-treated (n=4) and PBS-treated groups (n=4) gained weight. Body weights were significantly different (p<0.05) at days 10 and 11 (2-way repeated measures ANOVA, panel B). In rats fed the low-fat diet both groups gained weight; leptin-treated rats (n=4) gained significantly less (p<0.05) than PBS-treated rats (n=3) beginning at day 4 and lasting for the duration of the study (2-way repeated measures ANOVA, panel C). 118 Figure 6-6: IGF-1 Values Leptin treatment (n=3) significantly reduced IGF-1 in normal chow fed rats compared to PBS treatment (n=3) in the same diet group (p<0.05, 2-way ANOVA). Leptin treatment in the low-fat group (n=3) did not affect IGF-1 compared to PBS (n=2). In rats fed the high-fat diet, IGF-1 was attenuated in both the leptin(n=4) and PBS-treated (n=3) groups (p<0.05, 2-way ANOVA) compared to the other diets. Table 6-1: Organ Weights 119 Table 6-2: Body Temperature Discussion Food intake in rats fed the different diets, before leptin treatment, is reported. The normal chow rats ingested more grams of food than the other two groups for a majority of the study, but there were no differences in the intake of kcal among the three groups. As expected, the high-fat fed rats ingested the highest amount of fat while the low-fat rats ingested the lowest. These results resemble those seen in Chapter 3 in which the same diets were utilized. As was seen in Chapter 5, serum leptin levels increased over time in rats implanted with leptin-filled osmotic pumps indicating proper pump activity. In the normal chow and low-fat groups, the elevation of leptin was significant at week 2; although significance was not reached by week 1 there was an upward trend. In the highfat group, leptin was significantly elevated at weeks 1 and 2. Serum leptin levels were 120 not altered in PBS-implanted rats of any diet group over time. It would be expected that the serum leptin in the PBS-implanted high-fat diet groups would be elevated, however, beginning body weights were not the same prior to initiation of feeding. As seen in Chapter 5, steady state serum leptin concentrations were never achieved. In rats fed normal chow, both food intake and body weight were reduced beginning on days 2 and 3 of leptin administration, respectively. This effect was expected and has been well documented in other studies using leptin-filled osmotic pumps [Pelleymounter et al. 1995; Dawson et al. 1997]. By the end of the two-week implantation period, however, leptin lost its effects on food intake in the normal chow rats, indicating that leptin resistance was developing. When analyzed as grams of food ingested, leptin treatment enhanced food intake in rats fed the high-fat diet on days 7, 12, and 13. In addition, leptin had no effect on food intake in the low-fat group. When normalized to grams of food ingested per 100 grams body weight, however, these differential effects of leptin disappear. Leptin decreases food intake, as expected, in rats fed normal chow; however leptin is much less effective in rats fed the low-fat diet. In addition, leptin is virtually ineffective in rats fed the high-fat diet. Taken together, the results of leptin treatment on food intake in rats fed the different diets suggest that diets higher in calories (both the low-fat and the high-fat diets) inhibit leptin's effectiveness on this behavior. Body weight information was reported as change in body weight due to inconsistencies in weight at the time of implant. Leptin significantly attenuated gain in body weight in rats fed the low-fat diet for the greater part of the study. Recall that in normal chow rats, leptin acted to decrease body weight. In low-fat rats, leptin did not 121 decrease body weight, but prevented weight gain equivalent to that of the control rats fed the low-fat diet. Leptin had virtually no effect on body weight in rats fed the high-fat diet; body weight was attenuated in response to leptin, but only on days 10 and 11. Leptin regulated body weight more effectively in animals fed a low-fat diet than in animals fed a high-fat diet, but not as well as in rats fed normal chow. In the case of body weight, these results suggest diets higher in fat inhibit leptin's ability to curtail body weight. It may initially seem illogical that leptin can inhibit gain in body weight when there is no effect on food intake, as is seen in the low-fat diet group. However, factors other than those involved with feeding are implicated in the control of body weight. The rate of metabolism, for example, makes a considerable difference in body composition among individuals, and leptin has been shown to increase metabolism [Zhang et al. 1994; Campfield et al. 1995; Halaas et al. 1995; Pelleymounter et al. 1995; Halaas et al. 1997]. Also, there are direct effects of leptin to reduce the size of fat pads [Bai et al. 1996; Qian et al. 1998]. In the present study, there was no significant increase in body temperature in leptin-treated rats that would indicate an increased metabolism; however, since neither VO2max nor activity was measured, an increase in metabolism cannot be ruled out. The only conclusions that can be made regarding the metabolic status of the rats is related to IGF-1 levels. However, it was seen in Chapter 5 that the GH-regulating and metabolic effects of leptin are differentially regulated. In rats fed normal chow, leptin treatment decreased IGF-1 values, indicating that the stimulating effects of leptin on GH were reversed. Perhaps the mechanism by which resistance to the GH-regulating effects of leptin developed was a downregulation of 122 hypothalamic leptin receptor as was seen in the previous chapter. Also recall that the food intake-regulating effects of leptin were lost in these rats. In rats fed the high-fat diet, IGF-1 was suppressed in both leptin- and PBS-treated groups compared to either of the two other diets. Many investigators agree that leptin’s behavioral effects are attenuated in animals fed a highly palatable diet [Campfield et al. 1995; Frederich et al. 1995a; Masuzaki et al. 1995; Van Heek et al. 1997; Widdowson et al. 1997]. A study by Frederich’s lab [Frederich et al. 1995a] produced results similar to those reported in the current study: mice on a “Western” diet of 21% fat demonstrated elevated leptin levels. In Frederich's study, there was also an increase in body weight. The authors suggested that the high-fat diet acted to raise the physiological set-point for body weight. Another possibility exists: that elevated leptin levels produce leptin resistance causing the response to leptin to be attenuated. This phenomenon was shown in a study in which mice were fed a diet of 45% fat [Van Heek et al. 1997]. These mice experienced elevated serum leptin levels, grew obese, and were insensitive to peripherally administered leptin. In contrast to the resistance theory, Campfield et al. [1995] found that leptin, administered twice daily over two 5-day periods, continued to be effective at altering food intake and body weight in mice fed a high-fat diet. These results appear to argue against the development of leptin resistance, however perhaps resistance would have developed had the study been continued for a longer period of time or with continuous infusion via osmotic minipumps. Yet another possibility is that leptin's signal is overridden by the ingestion of a high-fat and/or high-calorie diet. 123 In the current study, we observed an attenuation of leptin's effects on food intake in animals fed diets high in calories (both low-fat and high-fat) and an attenuation of leptin's effects on body weight and GH secretion in animals fed a diet high in fat. These phenomena may be the result of an ability of a high-calorie or high-fat diet to impair leptin’s actions or to raise the body weight set-point defended by the hypothalamus, as was previously suggested [Frederich et al. 1995a]. Other possibilities exist in which the high-calorie or high-fat diet simply overrides or antagonizes leptin’s signal by stimulating other hypothalamic appetite systems [Frederich et al. 1995a] or downregulates leptin receptors in the blood-brain barrier or hypothalamus. Numerous "feeding peptides" are nutrient-specific. For example, neuropeptide Y (NPY) is well-known for its orexigenic effects, especially regarding the intake of carbohydrates [Stanley et al. 1985; Morley et al. 1987; Bray 1992a; Jhanwar-Uniyal et al. 1993; Wang et al. 1998b] and galanin specifically stimulates the ingestion of fat [Tempel et al. 1988; Bray 1992a]. Leptin has been shown to inhibit both NPY [Stephens et al. 1995; Schwartz et al. 1996c] and galanin [Sahu 1998] and as such, cannot be said to negatively regulate any specific type of nutrient. As a circulating factor informing the brain of the body’s energy stores, leptin plays an important role in the maintenance of body weight homeostasis. When the brain fails to recognize leptin, body weight homeostasis is impaired and obesity often develops. Obese animals and humans have elevated serum leptin levels and are insensitive to the effects of leptin [Frederich et al. 1995a; Maffei et al. 1995b; Halaas et al. 1997]. This socalled leptin resistance may be the result of one or more circumstances: inability of leptin to cross the blood-brain barrier [Banks et al. 1996; Caro et al. 1996a; Schwartz et al. 124 1996b; Van Heek et al. 1997], defective hypothalamic leptin receptors [Considine et al. 1996a ; Dawson et al. 1997], or impaired post-receptor signaling. The addition to this list of the ability of a high-fat or high-calorie diet to impair leptin’s actions or to alter body weight homeostasis regulated by the hypothalamus makes this an important area of study, especially in today's society where most diets are high in fat and where obesity is prevalent. In summary, leptin initially suppressed food intake and body weight in animals fed normal rat chow; but lost the effects on food intake and IGF-1 by week 2. Leptin inhibited body weight gain without altering food intake in rats fed a low-fat, high-calorie diet. In rats fed a high-fat, high-calorie diet, leptin had virtually no effect on food intake or body weight and IGF-1 was attenuated in both leptin- and PBS-treated subgroups in these rats. We conclude that leptin’s behavioral, metabolic, and GH-regulating effects are inhibited by the elevated intake of calories and fat indicating that animals that ingest such diets lose sensitivity to leptin. The results of this study further support the second hypothesis of this dissertation. CHAPTER 7 GENERAL DISCUSSION In the mammalian endocrine system, a variety of hormones and their hormonal and metabolic regulators are intricately intertwined, both centrally and in the periphery. Growth hormone (GH) and leptin are two such hormones. The synthesis of GH in and the release of GH from the anterior pituitary are stimulated and inhibited by two hypothalamic neuropeptides, GH-releasing hormone (GHRH) and somatotropin-release inhibiting hormone (SRIH), respectively. GH has many roles in the body, including the regulation of energy balance. Leptin, on the other hand, is secreted by adipocytes in proportion to fat mass. Leptin travels through the bloodstream to the brain where it informs the hypothalamus of the body's energy stores. The hypothalamus then regulates food intake and metabolic rate to maintain body weight homeostasis. Both GH and leptin are modulated by metabolic factors such as feeding, fasting, and obesity. GH and leptin also act to regulate each other, directly and/or indirectly. A natural feedback loop involving leptin and GH seems evident. In a normal physiological situation, the following occurs in the short-term feedback loop: leptin levels fall, food-seeking behavior is initiated, feeding occurs, leptin levels rise, and feeding is terminated. In this manner, leptin regulates food intake. In a longer-term feedback loop, if the effectiveness of leptin to regulate appetite is lost such as can occur in the presence of a diet high in calories and/or fat, animals may overeat, undergo hyperplasia and/or hypertrophy of fat cells, and consequently experience elevated levels of leptin. This 125 126 elevation in leptin may be sensed at the level of the hypothalamus and/or at the level of the pituitary to enhance GH secretion. GH stimulates lipolysis, which would then result in decreased or normalized leptin production. If the loss of effectiveness of leptin on food intake occurs chronically, leptin resistance develops and the regulation of GH by leptin may be lost. One of the hypotheses of this dissertation was that in the lean, nonleptin-resistant animal, circulating leptin stimulates the release of GH by acting indirectly at the level of the hypothalamus and/or directly at the level of the anterior pituitary. The studies completed in Chapters 3 and 4 support this hypothesis. In the case presented in Chapter 3, rats were given a high-fat or low-fat diet for 1 month. As expected, rats fed the high-fat diet gained more weight and secreted more leptin than controls. We consequently saw the predicted elevation in GH secretion, verified by elevated plasma IGF-1. There was no downregulation of leptin receptor mRNA in the hypothalamus, possibly indicating that no resistance had developed. The high fat content of the diet, however, prevent GH's lipolytic actions on body fat and normalization of leptin levels. This apparent loss of regulation of leptin by GH may have eventually resulted in the development of leptin resistance had the study been carried out longer. In fact, this development of resistance in animals fed the same diets over a longer period of time was seen in Chapter 6, and will be discussed in further detail later in this chapter. In Chapter 4, we sought to determine if leptin could produce its effects on GH directly at the level of the pituitary. A rat pituitary cell line was used in culture to eliminate indirect (hypothalamic) influence on GH production and secretion. We showed that leptin receptor mRNA is present in these cells and that leptin treatment significantly 127 increased GH secretion at 8 hours under appropriate conditions, in support of the original hypothesis. The effects of leptin on GH secretion was not significant at 24 hours, indicating that leptin lost some of its effectiveness over time. With continuous leptin treatment, leptin receptors on GH1 cells may be downregulated, resulting in a resistance to leptin. Leptin resistance had not developed in the studies completed in either Chapter 3 or 4, but in each study there was potential for the development of resistance. In the pathophysiological situation of leptin resistance, the hypothalamus loses the ability to detect circulating leptin and therefore continually suppresses GH secretion, as is seen in obesity. Leptin resistance can develop over time with a malfunction in distribution, delivery, metabolism, and/or elimination of leptin. It may also occur if leptin progressively loses its ability to cross the blood-brain barrier, perhaps with a downregulation of the transporter receptors (Ob-Ra) [Caro et al. 1996a]. Additionally, leptin resistance may occur with downregulation of the long-form of the receptor (Ob-Rb) or disruption of signal transduction. More than likely, there are multiple pathways involved in the development of leptin resistance. The second hypothesis of the dissertation is that in the leptin resistant state, leptin fails to stimulate GH. In obesity, GH is severely attenuated, and leptin resistance may be the one of the mechanisms by which this occurs. To support the second hypothesis, Chapter 5 analyzed leptin resistance as a result of hyperleptinemia due to exogenous leptin administration. Chapter 6 incorporated the special diets from Chapter 3 and the induced hyperleptinemia from Chapter 5 to further examine leptin resistance. 128 At least one mechanism by which the leptin resistance seen in Chapter 5 occurred was by a downregulation of hypothalamic leptin receptor mRNA and protein expression. It is a well-known pharmacological phenomenon that elevated levels of agonist, in this case circulating leptin, result in downregulation of receptor to prevent excessive activity. When this occurrence is chronic, there develops a state of resistance to the agonist. Leptin was infused continuously for 4 weeks via osmotic minipumps at one of two doses (0.1 mg/kg/day or 0.5 mg/kg/day). The rats in each treatment group became resistant to the long-term appetite regulating effects of leptin. In addition, both treatment groups were resistant to the leptin challenge at week 3. Interestingly, the control rats in the PBS group ate significantly more than the control rats in either leptin treatment group. These results suggest that, although resistance to the appetite-regulating effects of leptin was developing in the animals treated chronically, the long-term leptin treatment resulted in a ceiling effect where the rats could not eat as much as untreated rats. In addition, the leptin-treated animals were eating the same amount as the untreated animals at this time point. In contrast to the resistance to leptin on food intake, the animals in Chapter 5 did not become resistant to the body weight regulating effects of leptin. Our body weight and food intake results correspond to those from a study in which mice, after receiving daily leptin injections, experienced a normalization of the initially reduced food intake [Pelleymounter et al. 1995]. In that study, the reduction in body weight was never normalized. In addition, neither the high-dose nor the low-dose leptin-treated animals developed resistance to metabolic action of leptin as measured by serum insulin, glucose, 129 and triglyceride levels. These results indicate that leptin retained its ability to affect metabolic parameters even though its ability to alter food intake was lost. It was previously demonstrated in monosodium glutamate (MSG)-treated rats a dichotomy in leptin’s actions opposite to that seen in the current study. MSG-treated rats, with damage to the arcuate nucleus, retained sensitivity to the anorectic actions of leptin, but were resistant to its metabolic actions [Dawson et al. 1997]. The results of these two studies suggest that the food intake and metabolic effects of leptin are independently regulated. Chapter 6 combined the effects of the special diets and exogenously induced hyperleptinemia to investigate resistance. In Chapter 3, the diets were only fed for 1 month and resistance had not developed so in Chapter 6 the diets were fed for nearly 2 months before experimentation. Osmotic minipumps were implanted and PBS or leptin (0.25 mg/kg/day) was infused continuously for 2 weeks. Leptin levels were elevated in rats implanted with osmotic minipumps and also in rats fed the high-fat diet. In animals fed normal rat chow, leptin initially suppressed food intake and body weight, as would be expected. However, toward the end of the second week, food intake in the leptin-treated group was the same as in the PBS group. In addition, IGF-1 was attenuated, indicating that resistance to the food intake and GH-regulating effects of leptin had developed. In rats fed the low-fat diet, leptin inhibited body weight gain without altering food intake. Recall that the low-fat diet was high in calories. Perhaps the caloric content of the diet was responsible for the diet-induced resistance to the anorectic effects of leptin. There were no changes in IGF-1 in the low-fat rats. In rats fed the high-fat diet, leptin had virtually no effect on either body weight or food intake. IGF-1 was suppressed in both the leptin- and PBS-treated rats fed the high-fat diet. These results suggest that the 130 fat content of the diet was responsible for the apparent resistance of the GH-regulating effects of leptin. No metabolic indices were measured, however, so no statements regarding rate of metabolism would be substantiated. The effects of high-fat and/or high-calorie diets on leptin resistance have been observed previously [Campfield et al. 1995; Frederich et al. 1995a; Masuzaki et al. 1995; Van Heek et al. 1997; Widdowson et al. 1997]. The ability of a high-calorie or high-fat diet to impair leptin’s actions or to heighten the body weight set point preordained by the hypothalamus was previously suggested [Frederich et al. 1995a]. Other possibilities exist in which the high-calorie or high-fat diet simply overrides or antagonizes leptin’s signal by stimulating other hypothalamic appetite systems [Frederich et al. 1995a] or downregulates leptin receptors in the blood-brain barrier or hypothalamus. It is interesting to consider an opposing hypothesis. In this dissertation, it is hypothesized that in the normal physiological condition leptin stimulates GH and in the obese, leptin resistant condition leptin fails to stimulate GH. What if leptin were to inhibit GH in the physiological condition? Consider the case of a hibernating animal. During the warmer season, the animal must increase fat stores from which to live during hibernation. If the fat stores are increasing, leptin levels will also be increasing. It would be against the best survival mechanisms if GH were being stimulated and reducing fat stores in these animals. It would seem that, in this instance, the normal physiological function of leptin would be to reduce GH secretion. However, the physiology of a hibernating animal is unlike the physiology of a nonhibernating animal. Perhaps the effects of leptin in an animal preparing itself for hibernation oppose those in nonhibernating animals. As stated previously, as the animal gains weight in preparation 131 for hibernation and circulating leptin levels rise, it would be adverse for lipolysis to occur. Similarly, decrease of food intake and stimulation of metabolism would be harmful. The roles of leptin in these animals may be to inhibit GH, stimulate food intake, and reduce metabolic rate. Alternatively, leptin may have different roles at different times of year, perhaps under some seasonal circannual control. Yet another possibility is that the threshold at which leptin resistance develops is lowered in these animals. Consider another environmental phenomenon: starvation or famine. When food is scarce, animals are thin and circulating leptin levels would be low. Low leptin increases the drive to search for food and lowers metabolic rate to conserve energy. It has been shown that normal levels of leptin are required for normal GH secretion [Carro et al. 1997], hence in starvation GH is not stimulated by leptin. Both the instances of famine and hibernation can support the hypotheses proposed in this dissertation. This dissertation could continue in any of several directions. For example, additional studies on GH1 cells may include time-course and dose-response studies of leptin treatment. In addition, leptin receptor concentration could be measured under normal circumstances and after induction of leptin resistance. Furthermore, the same experiments could be completed in primary cultures of anterior pituitary cells. Additionally, supplementary diet studies could be completed in which the effects of various high-macronutrient diets (high-fat vs. high-carbohydrate vs. high-protein) are tested on leptin action. Summary As a circulating factor informing the brain of the body’s energy stores, leptin plays an important role in the maintenance of body weight homeostasis. When the brain 132 fails to recognize leptin, body weight homeostasis is impaired and obesity often develops. Obese animals and humans have elevated serum leptin levels and are insensitive to some of the effects of leptin [Frederich et al. 1995a; Maffei et al. 1995b; Halaas et al. 1997]. This so-called leptin resistance may be the result of one or more circumstances: inability of leptin to cross the blood-brain barrier [Banks et al. 1996; Caro et al. 1996a; Schwartz et al. 1996b; Van Heek et al. 1997], defective [Considine et al. 1996a ; Dawson et al. 1997] or downregulated hypothalamic leptin receptors, or impaired post-receptor signaling. The addition to this list of the ability of a high-fat or high-calorie diet to impair leptin’s actions or to heighten the body weight set point preordained by the hypothalamus makes this an important area of study, especially in today's society where most diets are high in fat and where obesity is prevalent. The feedback loop between leptin and GH helps maintain body weight homeostasis in the normal animal. We have demonstrated the development of resistance to the anorectic effects of leptin in conjunction with downregulation of hypothalamic leptin receptor. We have also shown that leptin’s behavioral and metabolic effects are inhibited by the elevated intake of calories and fat indicating that animals that ingest such diets lose sensitivity to leptin. We would like to suggest that when this feedback is disrupted as can occur with consumption of a high-fat diet, resistance to leptin can develop and obesity can develop. These results may be clinically important in the prevention and treatment of obesity and must be further explored in basic research. A possible model of leptin regulation of GH under normal and pathological conditions is given in Table 7-1. 133 Table 7-1: Model of GH Regulation by Leptin Physiological Potential Mechanisms ↓ leptin ↓GH Insufficient leptin to stimulate GH ↑ leptin ↑ GH Indirect action via hypothalamic GHRH, SRIH, NPY Direct action on anterior pituitary somatotropes Pathophysiological Potential Mechanisms ↑↑ leptin cannot ↑GH Leptin resistance Overriding of leptin action by high-fat and/or high-calorie diet High-fat and/or high-calorie diet elevation of hypothalamic body weight set-point REFERENCES Acs, Z., B. Szabo, G. Kapocs, and G.B. Makara. Gamma-aminobutyric acid stimulates pituitary growth hormone secretion in the neonatal rat: a superfusion study. Endocrinology 120:1790-1798, 1987. Ahima, R.S., D. Prabakaran, C. Mantzoros, D. Qu, B. Lowell, E. Maratos-Flier, and J.S. Flier. Role of leptin in the neuroendocrine response to fasting. Nature 382:250-252, 1996. Ahmad, I., J.A. Finkelstein, T.R. Downs, and L.A. Frohman. Obesity-associated decrease in growth hormone-releasing hormone gene expression: a mechanism for reduced growth hormone mRNA levels in genetically obese zucker rats. Neuroendocrinology 58:332-337, 1993. Ahren, B., S. Mansson, R.L. Gingerich, and P.J. Havel. Regulation of plasma leptin in mice: influence of age, high-fat diet, and fasting. Am. J. Physiol. 42:R113-R120, 1997. Almqvist, O., M. Thoren, M. Saaf, and O. Eriksson. Effects of growth hormone substitution on mental performance in adults with growth hormone deficiency: a pilot study. Psychoneuroendocrinology 11:347-352, 1986. Anand, B.K. and J.R. Brobeck. Hypothalamic control of food intake in rats and cats. Yale J. Biol. Med. 24:123-146, 1951. Argente, J., N. Caballo, V. Barrios, J. Pozo, M.T. Munoz, J.A. Chowden, and M. Hernandez. Mulitple endocrine abnormalities of the growth hormone and insulin-like growth factor axis in prepubertal children with exogenous obesity: effect of short- and long-term weight reduction. J. Clin. Endocrinol. Metab. 82:2076-2083, 1997. Argetsinger, L.S. and C. Carter-Su. Mechanism of signaling by growth hormone receptor. Physiol. Rev. 76:1089-1107, 1996. Argetsinger, L.S., G.W. Hsu, M.G. Myers, N. Billestrup, M.F. White, and C. Carter-Su. Growth hormone, interferon-gamma, and leukemia inhibitory factor promoted tyrosyl phosphorylation of insulin receptor substrate-1. J. Biol. Chem. 270:14685-14692, 1995. Auwerx, J. and B. Staels. Leptin. Lancet 351:737-742, 1998. Bado, A., S. Levasseur, S. Attoub, S. Kermorgant, J.-P. Laigneau, M.-N. Bortoluzzi, L. Moizo. T. Lehy, M. Guerre-Millo, Y. Le Marchand-Brustel, and M.J.M. Lewin. The stomach is a source of leptin. Nature 394:790-793, 1998. 134 135 Bai, Y., S. Zhang, K.-S. Kim, J.-K. Lee, and K.-H. Kim. Obese gene expression alters the ability of 30A5 preadipocytes to respond to lipogenic hormones. J. Biol. Chem. 271:13939-13942, 1996. Banks, W.A., A.J. Kastin, W. Huang, J.B. Jaspan, and L.M. Maness. Leptin enters the brain by a saturable system independent of insulin. Peptides 17:305-311, 1996. Barr, V.A., D. Malide, M.J. Zarnowski, S.I. Taylor, and S.W. Cushman. Insulin stimulates both leptin secretion and production by rat white adipose tissue. Endocrinology 138:4463-4472, 1997. Barzilai, N., J. Wang, D. Massilon, P. Vuguin, M. Hawkins, and L. Rossetti. Leptin selectively decreases visceral adiposity and enhances insulin action. J. Clin. Invest. 100:3105-3110, 1997. Baumann, G., K.D. Amburn, and T.A. Buchanan. The effect of circulating growthhormone-binding protein on metabolic clearance, distribution, and degradation of human growth hormone. J. Clin. Endocrinol. Metab. 64:657-660, 1987. Beck, B. and S. Richy. Hypothalamic hypocretin/orexin and neuropeptide Y: divergent interaction with energy depletion and leptin. Biochem. Biophys. Res. Comm. 258:119122, 1999. Bengtsson, B.A., R.J. Brummer, and I. Bosaeus. Growth hormone and body composition. Horm. Res. 33:19-24, 1990. Bengtsson, B.A., R.J. Brummer, S. Eden, I. Bosaeus, and G. Lindstedt. Body composition in acromegaly: the effect of treatment. Clin. Endocrinol. 31:481-190, 1989. Bengtsson, B.A., R.J. Brummer, S. Eden, T. Rosen, and L. Sjostrom. Effects of growth hormone on fat mass and fat distribution. Acta Paediatr. 383:62-65, 1992. Benitez, L., F. Mallo, C.V. Alvarez, B. Burguera, F. Sanchez-Franco, and C. Dieguez. Estrogen-dependent effects of bombesin on in vivo growth hormone secretion in the rat. Neuroendocrinology 52:608-611, 1990. Bennet, P.A., K. Lindell, C. Karlsson, I.C.A.F. Robinson, L.M.S. Carlsson, and B. Carlsson. Differential expression and regulation of leptin receptor isoforms in the rat brain: effects of fasting and oestrogen. Neuroendocrinology 67:29-36, 1998. Bennett, H.P. and C. McMartin. Peptide hormones and their analogues: distribution, clearance from the circulation, and inactivation in vivo. Pharmacol. Rev. 30:247-292, 1979. 136 Berneis, K., S. Vosmeer, and U. Keller. Rapid communication: effects of glucocorticoids and of growth hormone on serum leptin concentrations in man. Eur. J. Endocrinol. 135:663-665, 1996. Bernlohr, D.A., T.L. Doering, T.J. Kelly Jr., and M.D. Lane. Tissue specific expression of p422 protein, a putative lipid carrier, in mouse adipocytes. Biochem. Biophys. Res. Comm. 132:850-855, 1985. Beshyah, S.A., A. Henderson, R. Niththyanathan, P. Sharp, W. Richmond, and D.G. Johnston. Metabolic abnormalities in growth hormone deficient adults: carbohydrate tolerance and lipid metabolism. Endocrinol. Metab. 1:173-180, 1994. Bianda, T.L., Y. Glatz, M. Boeni-Schnetzler, E.R. Froesch, and C. Schmid. Effects of growth hormone (GH) and insulin-like growth factor-1 in GH-deficient adults. Diabetologia 40:363-364, 1997. Binnerts, A., G.R. Swart, J.H.P. Wilson, N. Hoogerbrugge, J.A.P. Pols, J.C. Birkenhager, and S.W.J. Lamberts. The effect of growth hormone administration in growth hormone deficient adults on bone, protein, carbohydrate and lipid homeostasis, as well as on body composition. Clin. Endocrinol. 37:79-87, 1992. Bjorbaek, C., J.K. Elmquist, P. Michl, R.S. Ahima, A. van Bueren, A.L. McCall, and J.S. Flier. Expression of leptin receptor isoforms in rat brain microvessels. Endocrinology 139:3485-3491, 1998. Bjorbaek C., S. Uotani, B. da Silva, and J.S. Flier. Divergent signaling capacities of the long and short isoforms of the leptin receptor. J. Biol. Chem. 272:32686-32695, 1997. Bjork, S., Jonsson, O. Westphal, and J.E. Levin. Quality of life of adults with growth hormone deficiency: a controlled study. Acta Pediatr. Scand. 356:55-59, 1989. Blum, W.F., M.B. Ranke, K. Kietzmann, E. Gauggel, H.J. Zeisel, and J.R. Bierich. A specific radioimmunoassay for growth hormone (GH)-dependent somatomedin binding protein: its use for diagnosis of GH deficiency. J. Clin. Endocrinol. Metab. 70:1292-1298, 1990. Boes, M., B.L. Dake, B.A. Booth, N.E. Erondu, Y. Oh, V. Hwa, R. Rosenfeld, and R.S. Bar. Connective tissue growth factor (IGFBP-rP2) expression and regulation in cultured bovine endothelial cells. Endocrinology 140:1575-1580, 1999. Boden, G., X. Chen, M. Mozzoli, and I. Ryan. Effect of fasting on serum leptin in normal human subjects. J. Clin. Endocrinol. Metab. 81:3419-3423, 1996. Borst, S.E., W.J. Millard, and D.T. Lowenthal. Growth hormone, exercise, and aging: the future of therapy for the frail elderly. J. Am. Ger. Soc. 42:528-535, 1994. 137 Bowers, C.Y. Editorial: on a peptidomimetic growth hormone-releasing peptide. J. Clin. Endocrinol. Metab. 79:940-942, 1994. Bowers, C.Y., F.A. Momany, D. Chang, A. Hong, and K. Chang. Structure-activity relationships of a synthetic pentapeptide that specifically releases GH in vitro. Endocrinology 106:663-667, 1980. Bowers, C.Y., F.A. Momany, A. Reynolds, and A. Hong. On the in vitro and in vivo activity of a new synthetic hexapeptide that acts on the pituitary to specifically release growth hormone. Endocrinology 114:1537-1545, 1984. Bowers, C.Y., A.O. Sartor, G.A. Reynolds, and T.M. Badger. On the actions of the growth hormone-releasing hexapeptide, GHRP. Endocrinology 128:2027-2035, 1991. Bray, G.A. Peptides affect the intake of specific nutrients and the sympathetic nervous system. Am. J. Clin. Nutr. 55:265S-271S, 1992a. Bray, G.A. Pathophysiology of obesity. Am. J. Clin. Nutr. 55:488S-494S, 1992b. Bray, G.A. and D.A. York. Hypothalamic and genetic obesity in experimental animals: an autonomic and endocrine hypothesis. Physiol. Reviews 59:719-809, 1979. Britton, D.R., G.F. Koob, J. Rivier, and W. Vale. Intraventricular corticotropin-releasing factor enhances behavioral effects of novelty. Life Sci. 31:363-367, 1982. Brobeck, J.R. Food intake as a mechanism of temperature regulation. Yale J. Biol. Med. 20:545-552, 1948. Brunani, A., C. Invitti, A. Dubini, R. Piccoletti, P. Bendinell, P. Maroni, G. Pezzoli, G. Ramella, A. Calogero, and F. Cavagnini. Cerebrospinal fluid and plasma concentrations of SRIH, beta-endorphin, CRH, NPY and GHRH in obese and normal weight subjects. Int. J. Obes. 19:17-21, 1995. Butkus, J.A., R.S. Brogan, A. Giustina, G. Kastello, M. Sothmann, and W.B. Wehrenberg. Changes in the growth hormone axis due to exercise training in male and female rats: secretory and molecular responses. Endocrinology 136:2664-2670, 1995. Cai, A. and J.F. Hyde. Upregulation of leptin receptor gene expression in the anterior pituitary of human growth hormone-releasing hormone transgenic mice. Endocrinology 139:420-423, 1998. Campfield, L.A., F.J. Smith, Y. Guisez, R. Devos, and P. Burn. Recombinant mouse OB protein: evidence for a peripheral signal linking adiposity and central neural networks. Science 269:546-549, 1995. 138 Caro, J.F., J.W. Kolacynski, M.R. Nyce, J.P. Ohannesian, I. Opentanova, W.H. Goldman, R.B. Lynn, P-L. Zhang, M.K. Sinha, and R.V. Considine. Decreased cerebrospinalfluid/serum leptin ratio in obesity: a possible mechanism for leptin resistance. Lancet 348:159-161, 1996a. Caro, J.F., M.K. Sinha, J.W. Kolaczynski, P.L. Zhang, and R.V. Considine. Leptin: the tale of the obesity gene. Diabetes 45:1455-1462, 1996b. Carro, E., R. Senaris, R.V. Considine, F.F. Casanueva, and C. Dieguez. Regulation of in vivo growth hormone secretion by leptin. Endocrinology 138:2203-2206, 1997. Carro, E., R.M. Senaris, L.M. Seoane, L.A. Frohman, A. Arimura, F.F. Casaneuva, and C. Dieguez. Role of growth hormone (GH)-releasing hormone and somatostatin on leptin-induced GH secretion. Neuroendocrinology 69:3-10, 1999. Carro, E., L.M. Seoane, R. Senaris, R.V. Considine, F.F. Casanueva, and C. Dieguez. Interaction between leptin and neuropeptide Y on in vivo growth hormone secretion. Neuroendocrinology 68:187-191, 1998. Casanueva, F.F., L. Villnueva, C. Dieguez, Y. Diaz, J.A. Cabranes, B. Szoke, M.F. Scanlon, A.V. Shally, and A. Fernandez-Cruz. Free fatty acids block growth hormone (GH) releasing-hormone GH secretion in man directly at the pituitary. J. Clin. Endocrinol. Metab. 65:634-642, 1987. Cavagnini, F., C. Invitti, and A. Di Landro. Effects of a GABA derivative, baclofen, on growth hormone and prolactin secretion in man. J. Clin. Endocrinol. Metab. 45:579-584, 1977. Chalew, S.A., R.A. Lozano, K.M. Armour, and A.A. Kowarski. Reduction of plasma insulin levels does not restore integrated concentration of growth hormone to normal in obese children. Int. J. Obes. 16:459-463, 1992. Chen, G., K. Koyama, X. Yuan, Y. Lee, Y.-T. Zhou, R. O-Doherty, C.B. Newgard, and R.H. Unger. Disappearance of body fat in normal rats induced by adenovirus-mediated leptin gene therapy. Proc. Natl. Acad. Sci. 93:14795-14799, 1996. Cheung, C.C., D.K. Clifton, and R.A. Steiner. Proopiomelanocortin neurons are direct targets for leptin in the hypothalamus. Endocrinology 138:4489-4492, 1997. Chihara, K., A. Arimura, and A.V. Schally. Effect of intraventricular injection of dopamine, norepinephrine, acetylcholine, and 5-hydroxytryptamine on immunoreactive somatostatin release into rat hypophyseal portal blood. Endocrinology 104:1656-1662, 1979. 139 Chronwall, B.M., D.A. DiMaggio, V.L. Massari, V.M. Pickel, D.A. Ruggiero, and T.L. O’Donohue. The anatomy of neuropeptide-Y-containing neurons in rat brain. Neuroscience 15:1159-1181, 1985. Chua, S.C., D.W. White, X.S. Wu-Peng, S.-M. Lu, N. Okada, E.E. Kershaw, W.K. Chung, L. Power-Kehoe, M. Chua, L.A. Tartaglia, and R.L. Leibel. Phenotype of fatty due to Gln269Pro mutation in the leptin receptor (Lepr). Diabetes 45:1141-1143, 1996. Cinti, S. Morphological and functional aspects of brown adipose tissue. Pediatr. Adolesc. Med. 2:125-132, 1992. Cioffi, J.A., A.W. Shafer, T.J. Zupancic, J. Smith-Gbur, A. Mikhail, D. Platika, and H.R. Snodgrass. Novel B219/OB receptor isoforms: possible role of leptin in hematopoiesis and reproduction. Nature Med. 2:585-589, 1996. Clark, J.T., P.S. Kalra, and S.P. Kalra. Neuropeptide Y stimulates feeding but inhibits sexual behavior in rats. Endocrinology 117:2435-2442, 1985. Clemmons, D.R. Variables controlling the secretion of a somatomedin-like peptide by cultured porcine smooth muscle cells. Circ. Res. 56:418-426, 1985. Clemmons, D.R. and D.S. Shaw. Variables controlling somatomedin production by cultured human fibroblasts. J. Cell. Physiol. 115:137-142, 1983. Clement, K., C. Garner, J. Hager, A. Philippi, C. LeDuc, A. Carey, T.J.R. Harris, C. Jury, L.R. Cardon, A. Basdevant, F. Dememais, B. Guy-Grand, M. North, and P. Froguel. Indication for linkage of the human ob gene region with extreme obesity. Diabetes 45:687-690, 1996. Clement, K., C. Vaisse, N. Lahlou, S. Cabrol, V. Pelloux, D. Casuto, M. Gourmelen, C. Dina, J. Chambaz, J.-M. Lacorte, A. Basdevant, P. Bougneres, Y. Lebouc, P. Froguel, and B. Guy-Grand. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 392:398-401, 1998. Cohen, B., D. Novick, and M. Rubinstein. Modulation of insulin activities by leptin. Science 274:1185-1188, 1996a. Cohen, S.L., J.L. Halaas, J.M. Friedman, and B.T. Chait. Human leptin characterization. Nature 382:589, 1996b. Coleman, D.L. Effects of parabiosis of obese with diabetes and normal mice. Diabetologia 9:294-298, 1973. Coleman, D.L. Obese and diabetes: two mutant genes causing diabetes-syndromes in mice. Diabetologia 41:141-148, 1978. 140 Coleman, D.L. and K.P. Hummel. Effects of parabiosis of normal with genetically diabetic mice. Am. J. Physiol. 217:1298-1304, 1969. Collins, S., K.W. Daniel, A.E. Petro, and R.S. Surwit. Strain specific responses to β3adrenergic receptor agonist treatment of diet-induced obesity in mice. Endocrinology 138:405-413, 1997. Collins, S., C.M. Kuhn, A.E.Petro, A.G. Swick, B.A. Chrunyk, and R.S. Surwit. Role of leptin in fat regulation. Nature 380:677, 1996. Commins, S.P., P.M. Watson, M.A. Padgett, A. Dudley, G. Argyropoulos, and T.W Gettys. Induction of uncoupling gene expression in brown and white adipose tissue by leptin. Endocrinology 140:292-300, 1999. Considine, R.V., E.L. Considine, C.J. Williams, T.M. Hyde, and J.F. Caro. The hypothalamic leptin receptor in humans: identification of incidental sequence polymorphisms and absence of the db/db mouse and fa/fa rat mutations. Diabetes 19:992994, 1996a. Considine, R.V., M.K. Sinha, M.L. Heiman, A. Kriauciunas, T.W. Stephens, M.R. Nyce, J.P. Ohannesian, C.C. Marco, L.J. McKee, T.L. Bauer, and J.F. Caro. Serumimmunoreactive leptin concentrations in normal weight and obese humans. N. Engl. J. Med. 334:292-295, 1996b. Cordido, F., F.F. Casanueva, and C. Dieguez. Cholinergic receptor activation by piridostigmine restores growth hormone (GH) responsiveness to GH-releasing hormone administration in obese subjects: evidence for blunted GH release of obesity. J. Clin. Endocrinol. Metab. 68:290-293, 1989. Cordido, F., A. Penalva, C. Dieguez, and F.F. Casanueva. Massive growth hormone (GH) discharge in obese subjects after the combined administration of GH-releasing hormone and GHRP-6: evidence for a marked somatotroph secretory capability in obesity. J. Clin. Endocrinol. Metab. 76:819-823, 1993. Cumin, F., H.-P. Baum, and N. Levens. Leptin is cleared from the circulation primarily by the kidney. Int. J. Obes. 20:1120-1126, 1996. Cuneo, R.C., F. Salomon, G.F. Watts, R. Hesp, and P.H. Sonksen. Growth hormone treatment improves serum lipids and lipoproteins in adults with growth hormone deficiency. Metabolism 42:1519-1523, 1993. Czernichow, P., M.C. Dauzet, M. Broyer, and R. Rappaport. Abnormal TSH, PRL and GH responses to TSH-releasing factor in chronic renal failure. J. Clin. Endocrinol. Metab. 43:630-637, 1976. Darnell, J.E.Jr. STATs and gene regulation. Science 277:1630-1635, 1997. 141 Darnell, J.E. Jr., I.M. Kerr, and G.R. Stark. JAK-STAT pathways and transcriptional activation in response to IFNs and other extracellular signaling proteins. Science 264:14151421, 1994. Davies, R.R., S.G. Turner, D. Cook, K.G.M.M. Alberti, and D.G. Johnston. The response of obese subjects to continuous infusion of human pancreatic growth hormone-releasing factor 1-44. 23:521-525, 1985. Dawson, C.M. and C.N. Hales. The effect of hypophysectomy on rat liver glucokinase activity and plasma glucose, insulin, and nonesterified fatty acid concentrations. Biochem. Biophys. Acta 184:287-298, 1969. Dawson, R. Jr., M.A. Pellymounter, W.J. Millard, S. Liu, and B. Eppler. Attenuation of leptin-mediated effects by monosodium glutamate (MSG)-induced arcuate nucleus damage. Am. J. Physiol. 273:E202-E206, 1997. DeBoer, H., G.J. Blok, H.J. Voerman, P.M.J.M. DeVries, and E.A. van der Veen. Body composition in adult growth hormone deficient men, assessed by anthropometry and bioimpedance analysis. J. Clin. Endocrinol. Metab. 75:833-837, 1992. Delitala, G., A. Grossman, and G.M. Besser. Opiate peptides control growth hormone through a cholinergic mechanism in man. Clin. Endocrinol. Oxf. 18:401-405, 1983. De Quiat, M.E. and P.C. Emson. Distribution of neuropeptide Y-like immunoreactivity in the rat central nervous system-II. Immunohistochemical analysis. Neuroscience 18:545618, 1986. Devos, R., J.G. Richards, L.A. Campfield, L.A. Tartaglia, Y. Guisez, J. van der Heyden, J. Travernier, G. Plaetinck, and P. Burn. OB protein binds specifically to the choroid plexus of mice and rats. Proc. Natl. Acad. Sci. 93:5668-5673, 1996. De Vos, P., R. Saladin, J. Auwerx, and B. Staels. Induction of ob gene expression by corticosteroids is accompanied by body weight loss and reduced food intake. J. Biol. Chem. 270:15958-15961, 1995. De Zegher, F., H. Devlieger, E. Eggermont, and J.D. Veldhuis. Properties of growth hormone and prolactin hypersecretion by the human infant on the day of birth. J. Clin. Endocrinol. Metab. 76:1177-1181, 1993. Dieguez, C. Leptin: a new GH secretagogue. [abstract]. International symposium on growth hormone basic aspects and new clinical applications. Lilly Press, Marbella, Spain, 1998. 142 Donaghue, K.C., T.M. Badger, W.J. Millard, L.S. Frisch, and W.E. Russell. Absence of ultradian rhythm or diurnal variation of circulating immunoreactive somatomedin-C in rats. Neuroendocrinology 52:1-8, 1990. Elias, C.F., C. Lee, J. Kelly, C. Aschkenasi, R.S. Ahima, P.R. Couceyro, M.J. Kuhar, C.B. Saper, and J.K. Elmquist. Leptin activates hypothalamic CART neurons projecting to the spinal cord. Neuron 21:1375-1385, 1998. Elmquist, J.K., R.S. Ahima, E. Maratos-Flier, J.S. Flier, and C.B. Saper. Leptin activates neurons in the ventrobasal hypothalamus and brainstem. Endocrinology 138:839-842, 1997. Elmquist, J.K., E. Maratos-Flier, C.B. Saper, and J.S. Flier. Unraveling the central nervous system pathways underlying responses to leptin. Nature Neurosci. 1:445-450, 1998. Emilsson, V., J.R.S. Arch, R.P de Groot, C.A. Lister, and M.A. Cawthorne. Leptin treatment increases suppressors of cytokine signaling in central and peripheral tissues. FEBS Lett. 455:170-174, 1999. Erickson, J.C., K.E. Clegg, and R.D. Palmiter. Sensitivity to leptin and susceptibility to seizures of mice lacking neuropeptide Y. Nature 381:415-418, 1996. Esler, M., M. Vaz, G. Collier, P. Nestel, G. Jennings, D. Kaye, D.Seals, and G. Lambert. Leptin in human plasma is derived in part from the brain, and cleared by the kidneys. Lancet 351:879, 1998. Escobar-Morreale, H.F, F.E. del Rey, and G.M. de Escobar. Thyroid hormones influence serum leptin concentrations in the rat. Endocrinology 138:4485-4488, 1997. Fagin, K.D., S.L. Lackey, C. Reagan, and M. DiGirolamo. Specific binding of growth hormone by rat adipocytes. Endocrinology 107:608-615, 1980. Fain, J.N., V.P. Kovacev, and R.O. Scow. Effect of growth hormone and dexamethasone on lipolysis and metabolism in isolated fat cells in the rat. J. Biol. Chem. 240:3522-3529, 1965. Farooqi, I.S., S.A. Jebb, G. Langmack, E. Lawrence, C.H. Cheetham, A.M. Prentice, I.A. Hughes, M.A. McCamish, and S. O'Rahilly. Effects of recombinant leptin therapy in a child with congenital leptin deficiency. N. Engl. J. Med. 341:879-884, 1999. Felsing, N.E., J.A. Brasel, and D.M. Cooper. Effect of low and high intensity exercise on circulating growth hormone in men. J. Clin. Endocrinol. Metab. 75:157-162, 1992. Fisker, S., N. Vahl, T.B. Hansen, J.O.L. Jorgensen, C. Hagen, H. Orskov, and J.S. Christiansen. Serum leptin is increased in growth hormone-deficient adults: relationship 143 to body composition and effects of placebo-controlled growth hormone therapy for 1 year. Metabolism 46:812-817, 1997. Fleury, C., M. Neverova, S. Collins, S. Raimbault, O. Champigny, C. Levi-Meyrueis, M.F. Seldin, R.S. Surwit, D. Ricquier, and C.H. Warden. Uncoupling protein-2: a novel gene linked to obesity and hyperinsulinemia. Nature Genet. 15:269-272, 1997. Flier, J.S. and J.K. Elmquist. Energetic pursuit of leptin function. Nature Biotechnol. 15:20-21, 1997. Florkowski, C.M., G.R. Collier, P.Z. Zimmet, J.H. Livesey, E.A. Espiner, and R.A. Donald. Low-dose growth hormone replacement lowers plasma leptin and fat stores without affecting body mass index in adults with growth hormone deficiency. Clin. Endocrinol. 45:769-773, 1996. Frederich, R.C., A. Hamann, S. Anderson, B. Lollmann, B.B.Lowell, and J.S. Flier. Leptin levels reflect body lipid content in mice: evidence for diet-induced resistance to leptin action. Nature Med. 1:1311-1314, 1995a. Frederich, R.C., B. Lollmann, A. Hamaan, A. Napolitano-Rosen, B.B. Kahn, B.B. Lowell, and J.S. Flier. Expression of ob mRNA and its encoded protein in rodents: impact of nutrition and obesity. J. Clin. Invest. 96:1658-1663, 1995b. Fulton, S., B. Woodside, and P. Shizgal. Modulation of brain reward circuitry by leptin. Science 287:125-128, 2000. Fuxe, K., L.F. Agnati, A. Harfstrand, P. Eneroth, A. Cintra, B. Tinner, E.M. Pich, M. Aronsson, B. Bunnemann, R. Lang, and D. Ganten. Studies of the neurochemical mechanisms underlying the neuroendocrine actions of neuropeptide Y. In: Neuropeptide Y, edited by V. Mutt, K. Fuxe, T. Hokfelt, and J.M. Lundberg, Raven Press, New York, 115-136, 1989. Ghigo, E., E. Arvat, G. Muccioli, and F. Camanni. Growth hormone-releasing peptides. Eur. J. Endocrinol. 136:445-460, 1997. Giustina, A., M. Doga, E. Bresciani, A.R. Bussi, L. Chiesa, V. Misitano, and G. Guistina. Effect of glucocorticoids on the paradoxical growth hormone response to thyrotropinreleasing hormone in patients with acromegaly. Metabolism 44:379-383, 1995. Giustina, A., A. Girelli, C. Bodini, C. Bonafanti, M. Doga, M. Schettino, and A. NegroVilar. Comparative effect of porcine and rat galanin on growth hormone secretion in normal adult men. Horm. Metab. Res. 24:90-91, 1992. Glass, A.R., K.D. Burman, W.T. Dahms, and T.M. Boehm. Endocrine function in human obesity. Metabolism 30:89-104, 1981. 144 Golay, A., A.L.M. Swislocki, Y.D.I. Chen, J.B. Jaspan, and G.M. Reaven. Effect of obesity on ambient plasma glucose, free fatty acid, insulin, growth hormone, and glucagon concentrations. J. Clin. Endocrinol. Metab. 63:481-484, 1986. Golden, P.L., T.J. Maccagnan, and W.M. Pardridge. Human blood-brain barrier leptin receptor. J. Clin. Invest. 99:14-18, 1997. Golstein, J., E. Van Cauter, D. Desir, P. Noel, J.P. Spire, S. Refetoff, and G. Copinschi. Effects of "jet-lag" on hormonal patterns: time shifts increase growth hormone release. J. Clin. Endocrinol. Metab. 56:433-440, 1983. Grant, M.B., B. Russell, C. Fitzgerald, and T.J. Merimee. Insulin-like growth factors in vitreous: studies in control and diabetic subjects with neovascularization. Diabetes 35:416-420, 1986. Grasso, P., M.C. Leinung, S.P. Unger, and D.W. Lee. In vivo effects of leptin-related synthetic peptides on body weight and food intake in female ob/ob mice: localization of leptin activity to domains between amino acid residues 106-140. Endocrinology 138:1413-1418, 1997. Green, H., M. Morikawa, and T. Nixon. A dual effector theory of growth-hormone action. Differentiation 29:195-198, 1985. Grundy, S.M. and J.P. Barnett. Disease-a-Month 36:645-696, 1990. Guerre-Millo, M. Regulation of ob gene overexpression in obesity. Biomed. Pharmacother. 51:318-323, 1997. Hakansson, M.-L., H. Brown, N. Ghilardi, R.C. Skoda, and B. Meister. Leptin receptor immunoreactivity in chemically defined target neurons of the hypothalamus. J. Neurosci. 18:559-572, 1998. Halaas, J.L., C. Boozer, J. Blair-West, N. Fidahusein, D.A. Denton, and J.M. Friedman. Physiological response to long-term peripheral and central leptin infusion in lean and obese mice. Proc. Natl. Acad. Sci. 94:8878-8883, 1997. Halaas, J.L., K.S. Gajiwala, M. Maffei, S.L. Cohen, B.T. Chait, D. Rabinowitz, R.L. Lallone, S.K. Burley, and J.M. Friedman. Weight-reducing effects of the plasma protein encoded by the obese gene. Science 269:543-546, 1995. Hall, K. and M. Bozovic. Stimulation of 35S incorporation into embryonic chick cartilage by extract from rat muscle. Horm. Metab. Res. 1:271-278, 1979. Han, P.W. Energy metabolism of tube-fed hypophysectomized rats bearing hypothalamic lesions. Am. J. Physiol. 215:1343-1350, 1968. 145 Hansen, T.B., J. Gram, P. Bjerre, C. Hagen, and J. Bollerslev. Body composition in active acromegaly during treatment with octreotide: a double-blind, placebo-controlled crossover study. Clin. Endocrinol. 41:323-329, 1994. Hansson, H.A., L.B. Dahlin, N. Danielsen, L. Fryklund, A.K. Nachemsson, P. Polleryd, B. Rozell, A. Skottner, S. Stemme, and G. Lundborg. Evidence indicating trophic importance of IGF-I in regenerating peripheral nerves. Acta Physiol. Scand. 126:609-614, 1986. Hansson, H.A., E. Jennische, and A. Skottner. Regenerating endothelial cells express insulin-like growth factor-I immunoreactivity after arterial injury. Cell Tissue Res. 250:499-505, 1987. Hardie, L.J., D.V. Rayner, S. Holmes, and P. Trayhurn. Circulating leptin levels are modulated by fasting, cold exposure and insulin administration in lean but not zucker (fa/fa) rats as measured by ELISA. Biochem. Biophys. Res. Comm. 223:660-665, 1996. Heiman, M.L., R.S. Ahima, L.S. Craft, B. Schoner, T.W. Stephens, and J.S. Flier. Leptin inhibition of the hypothalamic-pituitary-adrenal axis in response to stress. Endocrinology 138:3859-3863, 1997. Hetherington, A.W. and S.W. Ranson. Hypothalamic lesions and adiposity in the rat. Anat. Rec. 78:149-172, 1940. Heymsfield, S.B., A.S. Greenberg, K. Fujioka, R.M. Dixon, R. Kushner, T. Hunt, J.A. Lubina, J. Patane, B. Self, P. Hunt, and M. McCamish. Recombinant leptin for weight loss in obese and lean adults: a randomized, controlled, dose-escalation trial. JAMA 282:1568-1575, 1999. Hill, R.A., S. Margetic, G.G. Pegg, and C. Gazzola. Leptin: its pharmacokinetics and tissue distribution. Int. J. Obes. Relat. Metab. Disord. 22:765-770, 1998. Hilton, D., R. Richardson, W. Alexander, E. Viney, T. Willson, N. Sprigg, R. Starr, S. Nicholson, D. Metcalf, and N. Nicola. Twenty proteins containing a C-terminal SOCS box form five structural classes. Proc. Natl. Acad. Sci. 95:114-119, 1998. Ho, K.K., A.J. O'Sullivan, and D.M. Hoffman. Metabolic actions of growth hormone in man. Endocr. J. 43:S57-S63, 1996. Hochberg, Z., P. Hertz, B. Colin, S. Ish-Shalom, D. Yeshurun, M.B.H. Youdim, and T. Amit. The distal axis of growth hormone (GH) in nutritional disorders: GH-binding protein, insulin-like growth factor-1 (IGF-1), and IGF-1 receptors in obesity and anorexia nervosa. Metabolism 41:106-112, 1992. Hollenberg, A.N., V.S. Susulic, J.P. Madura, B. Zhang, D.E. Moller, P. Tontonoz, P. Sarraf, B.M Spiegelman, and B.B. Lowell. Functional antagonism between 146 CCAAT/Enhancer binding protein-alpha and peroxisome proliferator-activated receptorgamma on the leptin promoter. J. Biol. Chem. 272:5283-5290, 1997. Holmes, S.J., G. Economou, R.W. Whitehouse, J.E. Adams, and S.M. Shalet. Reduced bone mineral density in patients with adult onset growth hormone deficiency. J. Clin. Endocrinol. Metab. 78:669-674, 1994. Houben, H. and C. Denef. Effect of the bombesin receptor blockers [Leu13, psi CH2NHLeu]bombesinand N-pivaloyl GRP(20-25) alkyamide (L 686,095-001C002) on basal and neuromedin C-stimulated PRL and GH release in pituitary cell aggregates. Peptides 12:371-374, 1991. Houseknecht, K.L., C.S. Mantzoros, R. Kuliawat, E. Hadro, J.S. Flier, and B.B. Kahn. Evidence for leptin binding to proteins in serum of rodents and humans: modulation with obesity. Diabetes 45:1638-1643, 1996. Huang, Q., A. Viale, F. Picard, J.-L. Nahon, and D. Richard. Effects of leptin on melaninconcentrating hormone expression in the brain of lean and obese Lepob/Lepob mice. Neuroendocrinology 69:145-153, 1990. Hummel, K.P., M.M. Dickie, and D.L. Coleman. Diabetes, a new mutation in the mouse. Science 153:1127-1128, 1966. Jennische, E., A. Skottner, and H.A. Hansson. Satellite cells express the trophic factor IGF-I in regenerating skeleton muscle. Acta Physiol. Scand. 129:9-15, 1987. Ihle, J.N. and I.M. Kerr. JAKs and STATs in signaling by the cytokine receptor superfamily. Trends Genet. 11:69-74, 1995. Ihle, J.N., B.A. Witthuhn, F.W. Quelle, K. Yamamoto, and O. Silvennoinen. Signaling through the hematopoietic cytokine receptors. Annu. Rev. Immunol. 13:369-398, 1995. Iida, M., T. Murakami, M. Yamada, M. Sei, M. Kuwajima, A. Mizuno, Y. Noma, T. Aono, and K. Shima. Hyperleptinemia in chronic renal failure. Horm. Metab. Res. 28:724-727, 1996. Imaki, T., T. Shibasaki, K. Shizume, A. Masuda, M. Hotta, Y. Kiyosawa, K. Jibiki, H. Demura, T. Tsushima, and N. Ling. The effect of free fatty acids on growth hormone (GH)-releasing hormone-mediated GH secretion in man. J. Clin. Endocrinol. Metab. 60:290-293, 1985. Imura, H., I. Nakai, and T. Yoshimi. Effect of 5-hydrotryptophan on growth hormone and ACTH release in man. J. Clin. Endocrinol. Metab. 39:1-5, 1973. Ingalls, A.M., M.M. Dickie, and G.D. Snell. Obesity, a new mutation in the mouse. J. Hered. 41:317-318, 1950. 147 Iranmanesh, A., B. Grisso, and J.D. Veldhuis. Low basal and persistent pulsatile growth hormone secretion are revealed in normal and hyposomatotropic men studied with new ultrasensitive chemiluminescence assay. J. Clin. Endocrinol. Metab. 78:526-535, 1994. Iranmanesh, A., G. Lizarralde, and J.D. Veldhuis. Age and relative adiposity are specific negative dominants of the frequency and amplitude of growth hormone (GH) secretory bursts and the half-life of endogenous GH in healthy men. J. Clin. Endocrinol. Metab. 73:1081-1088, 1991. Jansen, G.R. and C.F. Hutchinson. Production of hypothalamic obesity by micro-surgery. Am. J. Physiol. 217:487-493, 1969. Jhanwar-Uniyal, M., B. Beck, Y.S. Jhanwar, C. Burlet, and S.F. Leibowitz. Neuropeptide Y projection from arcuate nucleus to parvocellular division of paraventricular nucleus: specific relation to the ingestion of carbohydrates. Brain Res. 631:97-106, 1993. Jin, L., B.G. Burguera, M.E. Couce, B.W. Scheithauer, J. Lamson, N.L. Eberhardt, E. Kulig, and R.V. Lloyd. Leptin and leptin receptor expression in the normal and neoplastic human pituitary: evidence of a regulatory role of leptin on pituitary cell proliferation. J. Clin. Endocrinol. Metab. 84:2903-2911, 1999. Jin, L., S. Zhang, B.G. Burguera, M.E. Couce, R.Y. Osamura, E. Kulig, and R.V. Lloyd. Leptin and leptin receptor expression in rat and mouse pituitary cells. Endocrinology 141:333-339, 2000. Johansson, J.O., J. Fowelin, K. Landin, I. Lager, and B.A. Bengtsson. Growth hormone deficient adults are insulin resistant. Metabolism 44:1126-1129, 1995. Johnston, J.A., L.M. Wang, E.P. Hanson, X.J. Sun, M.F. White, S.A. Oakes, J.H. Pierce, and J.J. O'Shea. Interleukins 2, 4, 7, and 15 stimulate tyrosine phosphorylation of insulin receptor substrates 1 and 2 in T cells. J. Biol. Chem. 270:28527-28530, 1995. Jorgensen, J.O., Pedersen, S.B., J. Borglum, N. Moller, O. Schmitz, J.S. Christiansen, and B. Richelsen. Fuel metabolism, energy expenditure, and thyroid function in growth hormone-treated obese women: a double-blind placebo controlled study. Metabolism 43:872-877, 1994. Kalra, S.P., J.T. Clark, A. Sahu, P.S. Kalra, and W.R. Crowley. Hypothalamic NPY: a local circuit in the control of reproduction and behavior. In: Neuropeptide Y, edited by V. Mutt, K. Fuxe, T. Hokfelt, and J.M. Lundberg, Raven Press, New York, 229-242, 1989. Kennedy, G.C. The hypothalamic control of food intake in rats. Proc. Roy. Soc. B 137:535549, 1950. 148 Kennedy, G.C. The role of depot fat in the hypothalamic control of food intake in the rat. Proc. Roy. Soc. B 140:578-592, 1953. Kieffer, T.J., R.S. Heller, and J.L. Habener. Leptin receptors expressed on pancreatic βcells. Biochem. Biophys. Res. Comm. 24:522-527, 1996. Kitajima, N., K. Chihara, H. Abe, Y. Okimura, Y. Fujii, M. Satoa, S. Shakutsui, M. Watanabe, and T. Fujita. Effects of dopamine on immunoreactive growth hormonereleasing factor and somatostatin secretion from rat hypothalamic slices perifused in vitro. Endocrinology 124:69-76, 1986. Kiyama, H. and P.C. Emson. Distribution of somatostatin mRNA in the rat nervous system as visualized by a novel non-radioactive in situ hybridization histochemistry procedure. Neuroscience 38:223-244, 1990. Klingenberg, M. Mechanism and evolution of the uncoupling protein of brown adipose tissue. Trends Pharmacol. Sci. 15:108-112, 1990. Knigge, U., B. Thuesen, A. Dejgaard, B. Svenstrup, and P. Bennett. Histamine-induced paradoxical GH response to TRH/GnRH in men and women: dependence on gonadal steroid hormones. Acta Endocrinol Copenh. 122:354-360, 1990. Koletsky, S. Obese spontaneously hypertensive rats - a model for the study of atherosclerosis. Exp. Mol. Pathol. 19:53-60, 1973. Kopelman, P.G. and K. Noonan. Growth hormone response to low dose intravenous injections of growth hormone releasing factor in obese and normal weight women. Clin. Endocrinol. 24:157-164, 1986. Klein, S., S.W. Coppack, V. Mohamed-Ali, and M. Landt. Adipose tissue leptin production and plasma leptin kinetics in humans. Diabetes 45:984-987, 1996. Kratzsch, J., B. Dehmel, F. Pulzer, E. Keller, P. Englaro, W.F. Blum, and M. Wabitsch. Increased serum GHBP levels in obese pubertal children and adolescents: relationship to body composition, leptin and indicators of metabolic disturbances. Int. J. Obes. 21:11301136, 1997. Kristensen, P., M.E. Judge, L. Thim, U. Ribel, K.N. Christiansen, B.S. Wulff, J.T. Clausen, P.J. Larsen, and S. Hastrup. Hypothalamic CART is a new anorectic peptide regulated by leptin. Nature 393:72-76, 1998. Lamson, G., L.C. Giudice, and R.G. Rosenfeld. Insulin-like growth factor binding proteins: structural and molecular relationship. Growth Factor 5:19-28, 1991. Lancranjan, I. and P. Marcack. New evidence for GH modulation by alpha-adrenergic system in man. Metabolism 26:1225-1229, 1977. 149 Lanzi, R., M.F. Manzoni, A.C. Andreotti, M.E. Malighetti, E. Bianchi, L. Piceni Sereni, A. Caumo, L. Luzi, and A.E. Pontiroli. Evidence for an inhibitory effect of physiological levels of insulin on the growth hormone (GH) response to GH-releasing hormone in healthy subjects. J. Clin. Endocrinol. Metab. 2:2239-2243, 1997. Lee, G-H., R. Proenca, J.M. Montez, K.M. Carroll, J.G. Darvishzadesh, J.I. Lee, and J.M. Friedman. Abnormal splicing of the leptin receptor in diabetic mice. Nature 379:632-635, 1996. Leibowitz, S.F. Hypothalamic neuropeptide Y and galanin: functional studies of coexistence with monoamines. In: Neuropeptide Y, edited by V. Mutt, K. Fuxe, T. Hokfelt, and J.M. Lundberg, Raven Press, New York, 267-282, 1989. Leung, D.W., S.A. Spencer, G. Cachianes, R.G. Hammonds, C. Collins, W.J. Henzel, R. Barnard, M.J. Waters, and W.I. Wood. Growth hormone receptor and serum binding protein: purification, cloning, and expression. Nature 330:537-543, 1987. Levin, N., C. Nelson, A. Gurney, R. Vandlen, and F. De Sauvage. Decreased food intake does not completely account for adiposity reduction after ob protein infusion. Proc. Natl. Acad. Sci. 93:1726-1730, 1996. Levine, J.A., N.L. Eberhardt, and M.D. Jensen. Role of nonexercise activity thermogenesis in resistance to fat gain in humans. Science 283:212-241, 1999. Licinio, J., C. Mantzoros, A.B. Negrao, G. Cizza, M-L. Wong, P.B. Bongiorno, G.P. Chrousos, B. Karp, C. Allen, J.S. Flier, and P.W. Gold. Human leptin levels are pulsatile and inversely related to pituitary-adrenal function. Nature Med. 3:575-579, 1997. Lieberman, S.A., A.G. Bjorkengren, and A.R. Hoffman. Rheumatological and skeletal changes in acromegaly. Endocrinol. Metab. Clin. North Am. 21:615-631, 1992. Liu, C., X-J. Liu, G. Barry, N. Ling, R.A. Maki, and E.B. de Souza. Expression and characterization of a putative high affinity human soluble leptin receptor. Endocrinology 138:3548-3554, 1997. Liuzzi, A., P.G. Chiodini, L. Botalla, G. Cremascoli, E.E. Mueller, and F. Silvestrini. Decreased plasma growth hormone (GH) levels in acromegalics following CB 154 (2-Bralpha-ergocryptide) administration. J. Clin. Endocrinol. Metab. 38:910-912, 1974. Locatelli, V., A. Torsello, M. Redaelli. E. Ghigo, F. Massara, F. Camanni, and E.E. Mueller. Cholinergic agonist and antagonist drugs modulate the growth hormone response to growth hormone-releasing hormone in the rat: evidence for mediation by somatostatin. J. Endocrinol. 11:271-278, 1986. 150 Lu, D., D. Willard, I.R. Patel, S. Kadwell, L. Overton, T. Kost, M. Luther, W. Chen, R.P. Woychik, W.O. Wilkison, and R.D. Cone. Agouti protein is an antagonist of the melanocyte-stimulating hormone receptor. Nature 371:799-802, 1994. Maccario, M., S. Grottoli, P. Razzore, M. Procopio, S.E. Oleandri, E. Ciccarelli, F. Camanni, and E. Ghigo. Effects of glucose load and/or arginine on insulin and growth hormone secretion in hyperprolactinemia and obesity. Eur. J. Endocrinol. 135:205-210, 1996. Maccario, M., M.R. Valetto, P. Savio, G. Aimaretti, C. Baffoni, M. Procopio, S. Grottoli, S.E. Oleandri, E. Avrat, and E. Ghigo. Maximal secretory capacity of somatotrope cells in obesity: comparison with GH deficiency. Int. J. Obes. Relat. Metab. Disord. 21:27-32, 1997. MacDougald, O.A., C.S. Hwang, H. Fan, and M.D. Lane. Regulated expression of the obese gene product (leptin) in white adipose tissue and 3T3-L1 adipocytes. Proc. Natl. Acad. Sci. 92:9034-9037, 1995. Maffei, M., H. Fei, G.-H. Lee, C. Dani, P. Leroy, Y. Zhang, R. Proenca, R. Negrel, G. Ailhaud, and J.M. Friedman. Increased expression in adipocytes of ob RNA in mice with lesions of the hypothalamus and with mutations at the db locus. Proc. Natl. Acad. Sci. 92:6957-6960, 1995a. Maffei, M., J. Halaas, E. Ravussin, R.E. Pratley, G.-H. Lee, Y. Zhang, H. Fei, S. Kim, R. Lallone, S. Ranganathan, P.A. Kern, and J.M. Friedman. Leptin levels in human and rodent: measurement of plasma leptin and ob RNA in obese and weight-reduced subjects. Nature Med. 1:1155-1161, 1995b. Martha Jr., P.M., A.D. Rogol, J.D Veldhuis, and R.M. Blizzard. A longitudinal assessment of hormonal and physical alterations during normal puberty in boys: the neuroendocrine growth axis during late prepuberty. J. Clin. Endocrinol. Metab. 81:40684074, 1996. Martin, J.B. Medical progress: neural regulation of growth hormone secretion. New. Engl. J. Med. 288:1384-1393, 1973. Martin, J.B. Twenty-third annual Bowditch lecture: brain mechanisms for the integration of growth hormone secretion. Physiologist 22:23-29, 1978. Martin, J.B., P. Brazeau, G.S. Tannenbaum, J.O. Willoughby, J. Epelbaum, L.C. Terry, and D. Durand. Neuroendocrine organization of growth hormone regulation. In: The Hypothalamus, volume 56. Eds: S. Reichlin, R. Baldessarini, and J.B. Martin, Raven Press, New York, 329-357, 1978. Martin, J.B. and W.J. Millard. Brain regulation of growth hormone secretion. J. Anim. Sci. 63:11-26, 1986. 151 Martin, R.J., D.J. Stolz, C.E. Allen, J.H. Gahagan, and W. Hymer. A comparison of pituitary cells from lean and obese zucker rats. Proc. Soc. Exp. Biol. Med. 172:8-10, 1983. Mason, G.A., G. Bissette, and C.B. Nemeroff. Effects of excitotoxic amino acids on pituitary hormone secretion in the rat. Brain Res. 289:366-369, 1983. Mason, M.M., Y. He, H. Chen, M.J. Quon, and M. Reitman. Regulation of leptin promoter function by Sp1, C-EBP, and a novel factor. Endocrinology 139:1013-1022, 1998. Masuzaki, H., Y. Ogawa, K. Hosoda, T. Kawada, T. Fushiki, and K. Nakao. Augmented expression of the obese gene in the adipose tissue from rats fed high-fat diet. Biochem. Biophys. Res. Comm. 216:355-358, 1995. Matsuda, J., I. Yokota, M. Iida, T. Murakami, E. Naito, M. Ito, K. Shima, and Y. Kuroda. Serum leptin concentration in cord blood: relationship to birth weight and gender. J. Clin. Endocrinol. Metab. 82:1642-1644, 1997. Matsuda, J., I. Yokota, Y. Tsuruo, T. Murakami, K. Ishimura, K. Shima, and Y. Kuroda. Developmental changes in long-form receptor expression and localization in rat brain. Endocrinology 140:5233-5238, 1999. Mauras, N., R.M. Blizzard, M.O. Thorner, and A.D. Rogol. Selective B1-adrenergic receptor-blockade with atenolol enhances basal and growth hormone releasing hormone mediated growth hormone release in man. Metabolism 36:369-372, 1987. Mauras, N., A.D. Rogol, M.W. haymond, and J.D. Veldhuis. Sex steroids, growth hormone, IGF-1: neuroendocrine and metabolic regulation in puberty. Horm. Res. 45:7480, 1996. Mayer, J. Glucostatic mechanism of regulation of food intake. N. Engl. J. Med. 249:1316, 1953. Mendelson, W.B., R.J.L.R. Wyatt, J.C. Gillin, and L.S. Jacobs. Piperidine enhances sleep-related and insulin-induced growth hormone secretion: further evidence for a cholinergic secretory mechanism. J. Clin. Endocinol. Metab. 52:409-415, 1981. McCann, S.M., V. Rettori, L. Milenkovic, M. Riedel, C. Augila, and J.K. McDonald. The role of neuropeptide Y (NPY) in control of anterior pituitary hormone release in the rat. In: Neuropeptide Y. Eds: V. Mutt, T. Hokfelt, K. Fuxe, and J.M. Lundberg, Raven Press, New York, 215-227, 1989. McDonald, K.A. Professor raises hackles with argument that obesity is not a health problem. The Chronicle of Higher Education, XLIII:A13-A14, 1996. 152 McGauley, G.A. Quality of life assessment before and after growth hormone treatment in adults with growth hormone deficiency. Acta Pediatr. Scand. 356:70-72, 1989. McGauley, G.A., R.C. Cuneo, F. Salomon, and P.H. Sonksen. Psychological well-being before and after growth hormone treatment in adults with growth hormone deficiency. Horm. Res. 33:52-54, 1990. McGregor, G.P., J.F. Desaga, K. Ehlenz, A. Fischer, F. Heese, A. Hegele, C. Lammer, C. Peiser, and R.E. Lang. Radioimmunological measurement of leptin in plasma of obese and diabetic human subjects. Endocrinology 137:1501-1504, 1996. McKay, L.D., N.J. Kenney, N.K. Edens, R.H. Williams, and S.C. Woods. Intracerebroventricular beta-endorphin increases food intake of rats. Life Sci. 29:14291434, 1981. Merabet, E., S. Dagogo-Jack, D.W. Coyne, S. Klein, J.V. Santiago, S.P. Hmiel, and M. Landt. Increased plasma leptin concentration in end-stage renal disease. J. Clin. Endocrinol. Metab. 82:847-850, 1997. Mercer, J.G., N. Hoggard, L.M. Williams, C.B. Lawrence, L.T. Hannah, and P. Trayhurn. Localization of leptin receptor mRNA and the long form splice variant (Ob-Rb) in mouse hypothalamus and adjacent brain regions by in situ hybridization. FEBS 387:113-116, 1996. Merimee, T.J. Growth hormone secretion and action. In: Endocrinology, volume 1. Ed: R. Degroot, Grune and Stratton, New York, 123-132, 1979. Miki, N., M. Ono, and K. Shizume. Evidence that opiatergic and alpha-adrenergic mechanisms stimulate rat growth hormone release via growth hormone-releasing factor (GRF). Endocrinology 114:1950-1952, 1984. Millard, W.J. Central regulation of growth hormone secretion. In: Animal Growth Regulation. Eds: Campion, D.R., G.J. Hausman, and R.J. Martin. Plenum Press, New York, 237-255, 1989. Millard, W.J., J.B. Martin, Jr., J. Audet, S.M. Sagar, and J.B. Martin. Evidence that reduced growth hormone secretion observed in monosodium glutamate-treated rats is the result of a deficiency in growth-hormone releasing factor. Endocrinology 110:540-550, 1981. Mizuno, T.M., H. Bergen, T. Funabashi, S.P. Kleopoulos, Y-G. Zhong, W.A. Bauman, and C.V. Mobbs. Obese gene expression: reduction by fasting and stimulation by insulin and glucose in lean mice, and persistant elevation in acquired (diet-induced) and genetic (yellow agouti) obesity. Proc. Natl. Acad. Sci. 93:3434-3438, 1996. 153 Mizuno, T.M. and C.V. Mobbs. Hypothalamic agouti-related protein messenger ribonucleic acid is inhibited by leptin and stimulated by fasting. Endocrinology 140:814817, 1999. Mohan, S. and D.J. Baylink. Insulin-like growth factor (IGF)-binding proteins in serum do they have additional roles besides modulating the endocrine IGF actions? J. Clin. Endocrinol. Metab. 81:3817-3820, 1996. Mokdad, A.H., M.K. Serdula, W.H. Dietz, B.A. Bowman, J.S. Marks, and J.P. Koplan. The spread of the obesity epidemic in the United States, 1991-1998. JAMA 282:15191522, 1999. Montague, C.T., I.S. Farooqi, J.P. Whitehead, M.A. Soos, H. Rau, N.J. Wareham, C.P. Sewter, J.E. Digby, S.N. Mohammed, J.A. Hurst, C.H. Cheetham, A.R. Earley, A.H. Barnett, J.B. Prins, and S. O’Rahilly. Congenital leptin deficiency is associated with severe early-onset obesity in humans. Nature 387:903-906, 1997. Morash, B., A. Li, P.R. Murphy, M. Wilkinson, and E. Ur. Leptin gene expression in the brain and pituitary gland. Endocrinology 140:5995-5998, 1999. Morley, J.E., A.S. Levine, B.A. Gosnell, J. Kneip, and M. Grace. Effect of neuropeptide Y on ingestive behaviors in the rat. Am. J. Physiol. 252:R599-R609, 1987. Mueller, E.E., A Panerai, D. Cocchi, I. Gil-Ad, G. Rossi, and V. Olgiati. Growth hormone releasing activity of thyrotropin-releasing hormone in rats with hypothalamic lesions. Endocrinology 100:1663-1667, 1977. Murakami, M., M. Narazaki, M. Hibi, H. Yawata, K. Yasukawa, M. Hamaguchi, T. Taga, and T. Kishimoto. Critical cytoplasmic region of the interleukin 6 signal transducer gp130 is conserved in the cytokine receptor family. Proc. Natl. Acad. Sci. 88:1134911353, 1991. Murakami, T., M. Iida, and K. Shima. Dexamethasone regulates obese expression in isolated rat adipocytes. Biochem. Biophys. Res. Comm. 214:1260-1267, 1995. Murakami, T., T. Yamashita, M. Iida, M. Juwajima, and K. Shima. A short form of the leptin receptor performs signal transduction. Biochem. Biophys. Res. Comm. 231:26-29, 1997. Murakami, Y., Y. Kato, Y. Kabayama, T. Inhoue, K. Kojo, H. Otha, and H. Imhura. Inhibition by antiserum to rat growth hormone release factor of growth hormone secretion induced by a met-enkephalin-analog, FK33 824, in rats. Proc. Soc. Exp. Biol. Med. 178:151-154, 1985. 154 Murakami, Y., Y. Kato, Y. Kabayama, K. Toyo, T. Inoue, and H. Imura. Involvement of growth hormone releasing factor in GH secretion induced by serotonergic mechanism in the conscious rat. Endocrinology 119:1089-1092, 1986. Murakami, Y., Y. Kato, A. Shimatsu, H. Koshiyama, N.Y. Hattori, N. Yanaihara, and H. Imura. Possible mechanisms involved in growth hormone secretion induced by galanin in the rat. Endocrinology 124:1224-1229, 1989. Murphy, J.E., S. Zhou, K. Giese, L.T. Williams, J.A. Escobedo, and V.J. Dwarki. Longterm correction of obesity and diabetes in genetically obese mice by a single intramuscular injection of recombinant adeno-associated virus encoding mouse leptin. Proc. Natl. Acad. Sci. 94:13921-13926, 1997. Murphy, L.J., K. Tach ibana, and H.G. Friesen. Stimulation of hepatic insulin-like growth factor-I gene expression by ovine prolactin: evidence for intrinsic somatogenic activity in the rat. Endocrinology 122:2027-2033, 1988. Must, A., J. Spadano, E.H. Coakley, A.E. Field, G. Colditz, and W.H. Dietz. The disease burden associated with overweight and obesity. JAMA 282:1523-1529, 1999. Nass, R., R.M. Huber, V. Klaus, O.A. Muller, J. Schopohl, and C. Strasburger. Effect of growth hormone (rGH) replacement therapy on physical work capacity and cardiac and pulmonary function in patients with rGH deficiency acquired in adulthood. J. Clin. Endocrinol. Metab. 80:552-557, 1995. National Institutes of Health Consensus Development Conference Statement. Health implications of obesity. Ann. Int. Med. 103:147-151, 1985. Netti, C., F. Guidobono, V.R. Olgiati, V. Sibila, and A. Pecile. Histamine agonist and antagonist drugs: interference with CNS control of GH release in rats. Horm. Res. 14:180-191, 1981. Nicholson, S. and D. Hilton. The SOCS proteins: a new family of negative regulators of signal transduction. J. Leukoc. Biol. 63:665-668, 1998. Okada, K., H. Sugihara, S. Minami, and I. Wakabayashi. Effect of parenteral administration of selected nutrients and central injection of antiserum to neuropeptide Y on growth hormone secretory pattern in food-deprived rats. Neuroendocrinology 57:678686, 1993. Ollmann, M.M., B.D. Wilson, Y.-K. Yang, J.A. Kerns, Y. Chen, I. Gantz, and G.S. Barsh. Antagonism of central melanocortin receptors in vitro and in vivo by agoutirelated protein. Science 278:135-138, 1997. Olney, J.W. Brain lesions, obesity, and other disturbances in mice treated with monosodium glutamate. Science 164:719-721, 1969. 155 Opie, L.H. and P.G. Walfish. Plasma free fatty acid concentration in obesity. N. Engl. J. Med. 268:757-760, 1963. Peillon, F., F. Cesselin, and B. Bression. In vitro effect of dopamine and L-dopa on prolactin and growth hormone release from human pituitary adenomas. J. Clin. Endocrinol. Metab. 49:737-741, 1979. Pelligrini, S. and I. Dusanter-Fourt. The structure, regulation and function of the Janus kinases (JAKs) and the signal transducers and activators of transcription (STATs). Eur. J. Biochem. 248:615-633, 1997. Pelleymounter, M.A., M.J. Cullen, M.B. Baker, R. Hecht, D. Winters, T. Boone, and F. Collins. Effects of the obese gene product on body weight regulation in ob/ob mice. Science 269:540-543, 1995. Philipps, A.F., K. Drakenberg, B. Persson, B. Sjogren, A.C. Eklof, K. Hall, and V.R. Sara. The effects of altered nutritional status upon insulin-like growth factor synthesis in neonatal rats. Pediatr. Res. 26:128-134, 1989. Poitout, V., C. Rouault, M. Guerre-Millo, I. Briaud, and G. Reach. Inhibition of insulin secretion by leptin in normal rodent islets of langerhans. Endocrinology 139:822-826, 1998. Polonsky, K.S., B.D. Biven, and E. van Cauter. Twenty-four-hour profiles and pulsatile patterns of insulin secretion in normal and obese subjects. J. Clin. Invest. 81:442-448, 1988. Pontiroli, A.E. and C. Scarpignato. Effect of bombesin on basal and stimulated secretion of some pituitary hormones in humans. Horm. Res. 23:129-135, 1986. Pralong, F.P., R. Roduit, G. Waeber, E. Castillo, F. Mosimann, B. Thorens, and R.C. Gaillard. Leptin inhibits directly glucocorticoid secretion by normal human and rat adrenal gland. Endocrinology 139:4264-4268, 1998. Qian, H., M.J. Azain, M.M. Compton, D.L. Hartzell, G.J. Hausman, and C.A. Baile. Brain administration of leptin causes deletion of adipocytes by apoptosis. Endocrinology 139:791-794, 1998. Qu, D., D.S. Ludwig, S. Gammeltoft, M. Piper, M.A. Pelleymounter, M.J. Cullen, W.F. Mathes, J. Przypek, R. Kanarek, and E. Maratos-Flier. A role for melanin-concentrating hormone in the central regulation of feeding behavior. Nature 380:243-247, 1996. Quabbe, H.J., M. Gregor, C. Bunke-Vogt, A. Eckhof, and J. Witt. Twenty-four hour pattern of growth hormone secretion in the rhesus monkey: studies including alterations in the sleep/wake cycle and sleep stage cycle. Endocrinology 109:513-522, 1981. 156 Quabbe, H.J. and U. Plockinger. Metabolic aspects of acromegaly and its treatment. Metabolism 45:61-62, 1996. Quintela, M. R. Senaris, H.L. Heiman, F.F. Casanueva, and C. Dieguez. Leptin inhibits in vitro hypothalamic somatostatin secretion and somatostatin mRNA levels. Endocrinology 138:5641-5644, 1997. Rasmussen, M.H., K.K.Y. Ho, L. Kjems, and J. Hilsted. Serum growth hormone-binding protein in obesity: effect of a short-term, very low calorie diet and diet-induced weight loss. J. Clin. Endocrinol. Metab. 81:1519-1524, 1996. Rau, H., B.J. Reaves, S. O'Rahilly, and J.P. Whitehead. Truncated human leptin (∇133) associated with extreme obesity undergoes proteasomal degradation after defective intracellular transport. Endocrinology 140:1718-1723, 1999. Ravussin, E. and E. Danforth, Jr. Beyond sloth - physical activity and weight gain. Science 283:184-185, 1999. Reed, D.R., Y. Ding, W. Xu, C. Cather, E.D. Green, and R.A. Price. Extreme obesity may be linked to markers flanking the human ob gene. Diabetes 45:691-694, 1996. Rettori, V., L. Milenkovic, M.C. Aguila, and S.M. McCann. Physiologically significant effect of neuropeptide Y to suppress growth hormone release by stimulating somatostatin discharge. Endocrinology 126:2296-2301, 1990. Reynolds, R.W. Ventromedial hypothalamic lesions without hyperphagia. Am. J. Physiol. 204:60-62, 1963. Richelsen, B., S.B. Pedersen, J.D. Borglum, T. Moller Pedersen, J. Jorgensen, and J.O. Jorgensen. Growth hormone treatment of obese women for 5 weeks: effect on body composition and adipose tissue LPL activity. Am. J. Physiol. 266:E211-E216, 1994. Roberts, C.T. Jr., S.R. Lasky, W.L. Lowe Jr., W.T. Seaman, and D. Leroith. Molecular cloning of rat insulin-like growth factor I complementary deoxyribonucleic acids: differential messenger ribonucleic acid processing and regulation by growth hormone in extrahepatic tissue. Mol. Endocrinol. 1:243-248, 1987. Rogers, Q.R. and P.M.B. Leung. The influence of amino acids on the neuroregulation of food intake. Fed. Proc. 32:1709-1719, 1973. Rohner-Jeanrenaud, F. and B. Jeanrenaud. Obesity, leptin, and the brain. N. Engl. J. Med. 334:324-325, 1996. 157 Rosen, T., L. Wiren, L. Wilhelmsen, I. Wiklund, and B.A. Bengtsson. Decreased psychological well-being in adult patients with growth hormone deficiency. Clin. Endocrinol. Oxf. 40:111-116, 1994. Russell-Jones, D.L., A.J. Weissberger. S.B. Bowes, J.M. Kelly, M. Thomason, A.M. Umpleby, R.H. Jones, and P.H. Sonksen. The effects of growth hormone on protein metabolism in adult growth hormone deficient patients. Clin. Endocrinol. Oxf. 38:427431, 1993. Sahu, A. Evidence suggesting that galanin (GAL), melanin-concentrating hormone (MCH), neurotensin (NT), proopiomelanocortin (POMC) and neuropeptide Y (NPY) are targets of leptin signaling in the hypothalamus. Endocrinology 139:795-798, 1998. Sakurai, T., A. Amemiya, M. Ishii, I. Matsuzaki, R.M. Chemelli, H. Tanaka, S.C. Williams, J.A. Richardson, G.P. Kozlowski, S. Wilson, J.R.S. Arch, R.E. Buckingham, A.C. Haynes, S.A. Carr, R.S. Annan, D.E. McNulty, W.-S. Liu, J.A. Terrett, N.A. Elshourbagy, D.J. Bergsma, and M. Yanagisawa. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92:573-585, 1998. Saladin, R., P. De Vos, M. Guerre-Millo, A. Leturque, J. Girard, B. Staels, and J. Auwerx. Transient increase in obese gene expression after food intake or insulin administration. Nature 377:527-529, 1995. Salomon, F., R.C. Cuneo, R. Hesp, J.F. Morris, L. Poston, and P.H. Sonksen. Basal metabolic rate in adults with growth hormone deficiency and in patients with acromegaly: relationship with lean body mass, plasma insulin level and leucocyte sodium pump activity. Clin. Sci. 83:325-330, 1992. Samson, W.K., T.C. Murphy, D. Robison, T.Vargas, E. Tau, and J.-K. Chang. A 35amino acid fragment of leptin inhibits feeding in the rat. Endocrinology 137:5182-5185, 1996. Sara, V.R. and K. Hall. Insulin-like growth factors and their binding proteins. Physiological Reviews 70:591-614, 1990. Sassolas, G. The role of Sandostatin in acromegaly. Metabolism 41:39-43, 1992. Scacchi, M., A. Pincelli, and F. Cavagnini. Growth hormone in obesity. Int. J. Obes. 23:260-271, 1999. Scarpace, P.J. and M. Matheny. Leptin induction of UPC1 gene expression is dependent on sympathetic innervation. Am. J. Physiol. 275:E259-E264, 1998. Scarpace, P.J., M. Matheny, B.H. Pollock, and N. Tumer. Leptin increases uncoupling expression and energy expenditure. Am. J. Physiol. 273:E226-E230, 1997. 158 Scapignato, C., F. Tirelli, and A.E. Pontiroli. Bombesin inhibits growth hormone response to insulin-induced hypoglycemia in humans. Brain Res. 371:187-189, 1986. Schindler, C. and J.E. Darnell Jr. Transcriptional responses to polypeptide ligands: the JAK-STAT pathway. Ann. Rev. Biochem. 64:621-651, 1995. Schober, E., H. Frisch, F. Waldhauser, and C. Bieglmayr. Influence of estrogen administration on growth hormone response to GHRH and L-Dopa in patients with Turner's syndrome. Acta Endocrinol. Copenh. 120:442-446, 1989. Schoenle, E., J. Zapf, R.E. Humbel, and E.R. Froesch. Insulin-like growth factor I stimulates growth in hypophysectomized rats. Nature 296:252-253, 1982. Schubring, C., W. Kiess, P. Englaro, W. Rascher, J. Dotsch, S. Hanitsch, A. Attanasio, and W.F. Blum. Levels of leptin in maternal serum, amniotic fluid, and arterial and venous cord blood: relation to neonatal and placental weight. J. Clin. Endocrinol. Metab. 82:1480-1483, 1997. Schwartz, M.W., D.G. Baskin, T.R. Bukowski, J.L. Kuijper, D. Foster, G. Lasser, D.E. Prunkard, D. Porte, Jr., S.C. Woods, R.J. Seely, and D.S. Weigle. Specificity of leptin action on elevated blood glucose levels and hypothalamic neuropeptide Y gene expression in ob/ob mice. Diabetes 45:531-535, 1996a. Schwartz, M.W., E. Peskind, M. Raskind, E.J. Boyko, and D. Porte. Cerebrospinal fluid leptin levels: relationship to plasma levels and to adiposity in humans. Nature Med. 2:589-593, 1996b. Schwartz, M.W., R.J. Seeley, L.A. Campfield, P. Burn, and D.G. Baskin. Identification of targets of leptin action in rat hypothalamus. J. Clin. Invest. 98:1101-1106, 1996c. Schwartz, M.W., R.J. Seeley, S.C. Woods, D.S. Weigle, L.A. Campfield, P. Burn, and D.G. Baskin. Leptin increases hypothalamic pro-opiomelanocortin mRNA expression in the rostral arcuate nucleus. Diabetes 46:2119-2123, 1997. Sclafani, A. and C.N. Berner. Hyperphagia and obesity produced by parasagittal and coronal hypothalamic knife cuts - further evidence for a longitudinal feeding inhibitory pathway. J. Comp. Physiol. 91:1000-1018, 1977. Senaris, R., T. Garcia-Caballero, X. Casabiell, R. Gallego, R. Castro, R.V. Considine, C. Dieguez, and F.F. Casanueva. Synthesis of leptin in human placenta. Endocrinology 138:4501-4504, 1997. Sharma, K., R.V. Considine, B. Michael, S.R. Dunn, L.S. Weisberg, B.R.C. Kurnik, P.B. Kurnik, J. O'Connor, M. Sinha, and J.F. Caro. Plasma leptin is partly cleared by the kidney and is elevated in hemodialysis patients. Kidney Int. 51:1980-1985, 1997. 159 Shibasaki, T., M. Hotta, A. Masuda, T. Imaki, N. Obara, and H. Demura. Plasma responses to GHRH and insulin-induced hypoglycemia in man. J. Clin. Endocrinol. Metab. 60:1265-1267, 1985. Shimabukuro, M., K. Koyama, G. Chen, M.-Y. Wang, F. Trieu, Y. Lee, C.B. Newgard, and R.H. Unger. Direct antidiabetic effect of leptin through triglyceride depletion of tissues. Proc. Natl. Acad. Sci. 94:4637-4641, 1997. Shuai, K., C. Schindler, V.R. Prezioso, and J.E. Darnell Jr. Activation of transcription by IFN-γ: tyrosine phosphorylation of a 91kDa DNA binding protein. Science 259:18081812, 1992. Shutter, J.R., M. Graham, A.C. Kinsey, S. Scully, R. Luthy, and K.L. Stark. Hypothalamic expression of ART, a novel gene related to agouti, is up-regulated in obese and diabetic mutant mice. Genes Dev. 11:593-602, 1997. Sinha, M.K. Human leptin: the hormone of adipose tissue. European J. of Endocrinol. 136:461-464, 1997. Sinha, M.K., J.P. Ohannesian, M.L. Heiman, A. Kriauciunas, T.W. Stephens, S. Magosin, C. Marco, and J.F. Caro. Nocturnal rise of leptin in lean, obese, and non-insulindependent diabetes mellitus subjects. J. Clin. Invest. 97:1344-1347, 1996a. Sinha, M.K., I. Opentanova, J.P. Ohannesian, J.W. Kolacznski, M.L. Heiman, J. Hale, G.W. Becker, R.R. Bowsher, T.W. Stephens, and J.F. Caro. Evidence of free and bound leptin in human circulation: studies in lean and obese subjects and during short-term fasting. J. Clin. Invest. 98:1277-1282, 1996b. Sinha, M.K., J. Sturis, J. Ohannesian, S. Magosin, T. Stephens, M.L. Heiman, K.S. Polonsky, and J.F. Caro. Ultradian oscillations of leptin secretion in humans. Biochem. Biophys. Res. Comm. 228:733-738, 1996c. Sivitz, W.I., S.A. Walsch, D.A. Morgan, M.J. Thomas, and W.G. Haynes. Effects of leptin on insulin sensitivity in normal rats. Endocrinology 138:3395-3401, 1997. Slieker, L.J., K.W. Sloop, P.L. Surface, A. Kriauciunas, F. LaQuier, J. Manetta, J. BueValleskey, and T.W. Stephens. Regulation and expression of ob mRNA and protein by glucocorticoids and cAMP. J. Biol. Chem. 271:5301-5304, 1996. Spiegelman, B.M. and J.S. Flier. Adipogenesis and obesity: rounding out the big picutre. Cell 87:377-389, 1996. Stanley, B.G., D.R. Daniel, A.S. Chin, and S.F. Leibowitz. Paraventricular nucleus injections of peptide YY and neuropeptide Y preferentially enhance carbohydrate ingestion. Peptides 6:1205-1211, 1985. 160 Starr, R. and D. Hilton. SOCS: suppressors of cytokine signaling. Int. J. Biochem. Cell Biol. 30:1081-1085, 1998. Starr, R., T.A. Willson, E.M. Viney, L.J. Murray, J.R. Rayner, B.J. Jenkins, T.J. Gonda, W.S. Alexander, D. Metcalf, N.A. Nicola, and D.J. Hilton. A family of cytokineinducible inhibitors of signaling. Nature 387:917-921, 1997. Stephens, T.W., M. Basinski, P.K. Bristow, J.M. Bue-Valleskey, S.G. Burgett, L. Craft, J. Hale, J. Hoffmann, H.M. Hsiung, A. Kriauciunas, W. MacKellar, P.R. Rosteck, Jr., B. Schoner, D. Smith, F.C. Tinsley, X.-Y. Zhang, and M. Heiman. The role of neuropeptide Y in the antiobesity action of the obese gene product. Nature 377:530-532, 1995. Steiner, R.A. Editorial: lords and ladies leapin' on leptin. Endocrinology 137:4533-4534, 1996. Steiner, R.A., J.K. Stewart, J. Barber, D. Koerker, C.J. Goodner, A. Brown, P. Illner, and C.C. Gale. Somatostatin: a physiological role in the regulation of growth hormone secretion in the adolescent male baboon. Endocrinology 102:1587-1594, 1978. Strobel, A., T. Issad, L. Camoin, M. Ozata, and A.D. Strosberg. A leptin missense mutation associated with hypogonadism and morbid obesity. Nature Genet. 18:213-215, 1998. Takaya, K., Y. Ogawa, J. Hiraoka, K. Hosoda, Y. Yamori, and K. Nakao. Nonsense mutation of leptin receptor in the obese spontaneously hypertensive Koletsky rat. Nature Genet. 14:130-131, 1996. Tanizawa Y., S. Okuya, H. Ishihara, T. Asano, T. Yada, and Y. Oka. Direct stimulation of basal insulin secretion by physiological concentrations of leptin in pancreatic β-cells. Endocrinology 138:4513-4516, 1997. Tannenbaum, G.S. Multiple levels of cross-talk between somatostatin (SRIF) and growth hormone (GH)-releasing factor in genesis of pulsatile GH secretion. Clin. Pediatr. Endocrinol. 3[suppl5]:97-110, 1994. Tannenbaum, G.S., E. Colle, W. Gurd, and L. Wanamaker. Dynamic time-course studies of the spontaneously diabetic BB wistar rat. I. Longitudinal profiles of plasma growth hormone, insulin, and glucose. Endocrinology 109:1872-1879, 1981. Tannenbaum, G.S., W. Gurd, and M. Lapointe. Leptin is a potent stimulator of spontaneous pulsatile growth hormone (GH) secretion and the GH response to GHreleasing hormone. Endocrinology 139:3871-3875, 1998. 161 Tannenbaum, G.S., M. Lapointe, W. Gurd, and J.A. Finkelstein. Mechanisms of impaired growth hormone secretion in genetically obese Zucker rats: roles of growth hormonereleasing factor and somatostatin. Endocrinology 127:3087-3095, 1990. Tannenbaum, G.S. and N. Ling. The interrelationship of growth hormone (GH)-releasing factor and somatostatin in generation of the ultradian rhythm of GH secretion. Endocrinology 115:1952-1957, 1984. Tartaglia, L.A. The leptin receptor. J. Biol. Chem. 272:6093-6096, 1997. Tartaglia, L.A., M. Dembski, X. Weng, N. Deng, J. Culpepper, R. Devos, G.J. Richards, L.A. Campfield, F.T. Clark, J. Deeds, C. Muir, S. Sanker, A. Moiarty, K.J. Moore, J.S. Smuto, G.G. Mays, E.A. Woolf, C.A. Monroe, and R.I. Tepper. Identification and expression cloning of a leptin receptor, OB-R. Cell 83:1263-1271, 1995. Tatemoto, K. Neuropeptide Y: isolation, structure and function. In: Neuropeptide Y. Eds: Mutt, V., K. Fuxe, T. Hokfelt, and J.M. Lundberg, Raven Press, New York, 13-22, 1989. Tatemoto, K., M. Carlquist, and V. Mutt. Neuropeptide Y - a novel brain peptide with structural similarities to peptide YY and pancreatic polypeptide. Nature 296:659-660, 1982. Tempel, D.L., K.J. Leibowitz, and S.F. Leibowitz. Effects of PVN galanin on macronutrient selection. Peptides 9:309-314, 1988. Thornton, J.E., C.C. Cheung, D.K. Clifton, and R.A. Steiner. Regulation of hypothalamic proopiomelanocortin mRNA by leptin in ob/ob mice. Endocrinology 138:5063-5066, 1997. Tollet-Egnell, P., A. Flores-Morales, A. Stavreus-Evers, L. Sahlin, and G. Norstedt. Growth hormone regulation of SOCS-2, SOCS-3, and CIS messenger ribonucleic acid expression in the rat. Endocrinology 140:3693-3704, 1999. Truett, G.E., N. Bahary, J.M. Friedman, and R.L. Leibel. Rat obesity gene fatty (fa) maps to chromosome 5: evidence for homology with the mouse gene diabetes (db). Proc. Natl. Acad. Sci. 88:7806-7809, 1991. Uotani, S., C. Bjorbaek, J. Tornoe, and J.S. Flier. Functional properties of leptin receptor isoforms: internalization and degradation of leptin and ligand-induced receptor downregulation. Diabetes 48:279-286, 1999. Vaisse, C., J.L. Halaas, C.M. Horvath, J.E. Darnell, Jr., M. Stoffel, and J.M. Friedman. Leptin activation of stat3 in the hypothalamus of wild-type and ob/ob mice but not db/db mice. Nature Genet. 14:95-97, 1996. 162 Valentini, U., A. Cimino, A. Rotondi, R. Pelizzari, A. Giustina, C. Marchetti, and G. Romanelli. Growth hormone response to thyrotropin releasing hormone and placebo in a group of insulin-dependent diabetic patients. J. Endocrinol. Invest. 12:643-646, 1989. Van Heek, M., D.S. Compton, C.F. France, R.P. Tedesco, A.B. Fawzi, M.P. Graziano, E.J. Sybertz, C.D. Strader, and H.R. Davis, Jr. Diet-induced obese mice develop peripheral, but not central, resistance to leptin. J. Clin. Invest. 99:385-390, 1997. Veldhuis, J.D. and A. Iranmanesh. Physiological regulation of the human growth hormone (GH)-insulin-like growth factor type I (IGF-I) axis: predominant impact of age, obesity, gonadal function, and sleep. Sleep 19:S221-S224, 1996. Veldhuis, J.D. and M.L. Johnson. Cluster analysis: a simple, versatile, and robust algorithm for endocrine pulse detection. Am. J. Physiol. 250:E486-E493, 1986. Veldhuis, J.D., A. Iranmanesh, K.K.Y. Ho, M.J. Waters, M.L. Johnson, and G. Lizarralde. Dual defects in pulsatile growth hormone secretion and clearance subserve the hyposomatotropism of obesity in man. J. Clin. Endocrinol. Metab. 72:51-58, 1991. Veldhuis, J.D., A. Iranmanesh, A.D. Rogol, and R.J. Urban. Regulatory actions of testosterone on pulsatile growth hormone secretion in the human: studies using deconvolution analysis. In: Somatotropic Axis and the Reproductive Process in Health and Disease. Eds: E.Y. Adashi and M.O. Thorner, Springer-Verlag, New York, 40-57, 1995a. Veldhuis, J.D., A.Y. Liem, S. South, A. Weltman, J. Weltman, D.A. Clemmens, R. Abbott, T. Mulligan, M.L. Johnson, S. Pincus, M. Straume, and A. Iranmanesh. Differential impact of age, sex, steroid hormones, and obesity in basal versus pulsatile growth hormone secretion in men as assessed in an ultrasensitive chemiluminescence assay. J. Clin. Endocrinol. Metab. 80:3209-3222, 1995b. Vikman, K., B. Carlsson, H. Billig, and S. Eden. Expression and regulation of growth hormone (GH) receptor messenger ribonucleic acid (mRNA) in rat adipose tissue, adipocytes, and adipocyte precursor cells: GH regulation of GH receptor mRNA. Endocrinology 129:1155-1161, 1991. Vila, R., C. Adan, I. Rafecas, J.A. Fernandez-Lopez, X. Remesar, and M. Alemany. Plasma leptin turnover rates in obese zucker rats. Endocrinology 139:4466-4469, 1998. Vincent, S.R., L. Skirboll, T. Hokfelt, O. Johansson, J.M. Lundberg, R.P. Elde, L. Terenius, and J. Kimmel. Coexistence of somatostatin- and avian pancreatic polypeptide (APP)-like immunoreactivity in some forebrain neurons. Neuroscience 7:439-446, 1982. Vuagnat, B.A.M., D.D. Pierroz, M. Lalaoui, P. Englaro, F.P. Pralong, W.F. Blum, and M.L. Aubert. Evidence for a leptin-neuropeptide Y axis for the regulation of growth hormone secretion in the rat. Neuroendocrinology 67:291-300, 1998. 163 Wabitsch, M., P.B. Jensen, W.F. Blum, C.T. Christoffersen, P. Englaro, E. Heinze, W. Rascher, W. Teller, J. Tornqvist, and H. Hauner. Insulin and cortisol promote leptin production in cultured human fat cells. Diabetes 45:1435-1438, 1996. Wang, D.H., K. Ishii, E. Seno, S. Yane, T. Horike, H. Yamamoto, N. Suganuma, M. Arimichi, and K. Taketa. Reduced serum levels of ALT and GGT and high carbohydrate intake among workers exposed to toluene below the threshold limit values. Ind. Health 36:14-19, 1998a. Wang J., A. Akabayashi, J. Dourmashkin, H.J. Yu, J.T. Alexander, H.J. Chae, and S.F. Leibowitz. Neuropeptide Y in relation to carbohydrate intake, corticosterone and dietary obesity. Brain Res. 802:75-88, 1998b. Wang, J., R. Liu, M. Hawkins, N. Barzilai, and L. Rossetti. A nutrient-sensing pathway regulates leptin gene expression in muscle and fat. Nature 393:684-688, 1998c. Wang, M.-Y., Y.T. Zhou, C.B. Newgard, and R.H. Unger. A novel leptin receptor isoform in rat. FEBS Letters 392:87-90, 1996. Wang, Y.D., K. Wong, and W.I. Wood. Intracellular tyrosine residues of the human growth hormone receptor are not required for the signaling of proliferation or Jak-STAT activation. J. Biol. Chem. 270:7021-7024, 1995. Weil, R. Pituitary growth hormone and intermediary metabolism: the hormonal effect on the metabolism of fat and carbohydrate. Acta Endocrinol. Copenh. 49:7-92, 1965. White, D.W., K.K. Kuropatwinski, R. Devos, H. Baumann, and L.A. Tartaglia. Leptin receptor (Ob-R) signaling: cytoplasmic domain mutational analysis and evidence for receptor homo-oligomerization. J. Biol. Chem. 272:4065-4071, 1997. White, D.W. and L.A. Tartaglia. Evidence for ligand-independent homo-oligomerization of leptin receptor (Ob-R) isoforms: a proposed mechanism permitting productive longform signaling in the presence of excess short-form expression. J. Cell. Biochem. 73:278288, 1999. Widdowson, E.M., O.G. Edholm, and R.A. McCance. The food intake and energy expenditure of cadets in training. Br. J. Nutr. 8:147-155, 1954. Widdowson, P.S., R. Upton, R. Buckingham, J. Arch, and G. Williams. Inhibition of food response to intracerebroventricular injection of leptin is attenuated with diet-induced obesity. Diabetes 46:1782-1785, 1997. Wiesner, G., M. Vaz, G. Collier, D. Seals, D. Kaye, G. Jennings, G. Lambert, D. Wilkinson, and M. Esler. Leptin is released from the human brain: influence of adiposity and gender. J. Clin. Endocrinol. Metab. 84:2270-2274, 1999. 164 Wilding, J. A weighty problem. Nature 391:759, 1998. Wilding, J.P.H., S.G. Gilbey, C.J. Bailey, R.A.L. Batt, G. Williams, M.A. Ghatei, and S.R. Bloom. Increased neuropeptide–Y messenger ribonucleic acid (mRNA) and decreased neurotensin mRNA in the hypothalamus of the obese (ob/ob) mouse. Endocrinology 132:1939-1944, 1993. Williams, T., M. Berelowitz, S.N. Joffe, M.O. Thorner, J. Rivier, W. Vale, and L.A. Frohman. Impaired growth hormone responses to growth hormone-releasing factor in obesity. N. Engl. J. Med. 311:1403-1407, 1984. Wilson, B.D., D. Bagnol, C.B. Kaelin, M.M. Ollmann, I. Gantz, S.J. Watson, and G.S. Barsh. Physiological and anatomical circuitry between agouti-related protein and leptin signaling. Endocrinology 140:2387-2397, 1999. Winston, L.A. and T. Hunter. Intracellular signaling: putting JAKs on the kinase MAP. Curr. Biol. 6:668-671, 1996. Woods, A.J. and M.J. Stock. Leptin activation in hypothalamus. Nature 381:745, 1996. Wu-Peng, X.S., S.C. Chua Jr., N. Okada, S.M. Liu, M. Nicholson, and R.L. Leibel. Phenotype of the obese Koletsky (f) rat due to Tyr763Stop mutation in the extracellular domain of the leptin receptor (Lepr): evidence for deficient plasma-to-CSF transport of leptin in both Zucker and Koletsky obese rat. Diabetes 46:513-518, 1997. Yamashita, T., T. Murakami, S. Otani, M. Kuwajima, and K. Shima. Leptin receptor signal transduction: OBRa and OBRb of fa type. Biochem. Biophys. Res. Comm. 246:752-759, 1998. Yoshida, T., M. Hayashi, T. Monkawa, and T. Saruta. Regulation of obese mRNA expression by hormonal factors in primary cultures of rat adipocytes. Eur. J. Endocrinol. 135:619-625, 1996. Yoshimura, A., T. Ohkubo, T. Kiguchi, N.A. Jenkins, D.J. Gilbert, N.G. Copeland, T. Hara, and A. Miyajima. EMBO J. 14:2816-2826, 1995. Zamorano, P.L., V.B. Mahesh, L.M. De Sevilla, G.K. Bhat, and D.W. Brann. Expression and localization of the leptin receptor in endocrine and neuroendocrine tissues of the rat. Neuroendocrinology 65:223-228, 1997. Zhang, F., M.B. Basinski, J.M. Beals, S.L. Briggs, L.M. Churgay, D.K. Clawson, R.D. DiMarchi, T.C. Furman, J.E. Hale, H.M. Hsiung, B.E. Schoner, D.P. Smith, X.Y. Zhang, J.-P. Wery, and R.W. Schevitz. Crystal structure of the obese protein leptin-E100. Nature 387:206-209, 1997. 165 Zhang, Y., R. Proenca, M. Maffei, M. Barone, L. Leopold, and J.M. Friedman. Positional cloning of the mouse obese gene and its human homologue. Nature 372:425-432, 1994. Zhau, J., L. Unelius, T. Bengtsson, B. Cannon, and J. Nedergaard. Coexisting βadrenoceptor subtypes: significance for thermogenic process in brown fat cells. Am. J. Physiol. 267:C969-C979, 1994. Zucker, L.M. and T.F. Zucker. Fatty, a new mutation in the rat. J. Hered. 52:275278,1961. Zurlo, F., R.T. Ferraro, A.M. Fontvielle, R. Rising, C. Bogardus, and E. Ravussin. Spontaneous physical activity and obesity: cross-sectional and longitudinal studies in Pima Indians. Am. J. Physiol. 263:E296-E300, 1992. BIOGRAPHICAL SKETCH Robin Leigh Picking was born in Crestline, Ohio, on February 20, 1968 to Thomas R. and Nicolle A. Picking and older brother, T. Reed Picking (age 4). When Robin was 11, the family moved to Clearwater, Florida, where she attended John F. Kennedy Middle School and Clearwater High School. Like any teenager, Robin was anxious to leave the nest and go far from home, so she enrolled in Newberry College in South Carolina. The plan to get away backfired, however, when Robin met her future husband, George Martin, during her first summer break back in Clearwater. Now anxious to return to Clearwater to be with George, Robin worked hard and finished college one semester early, graduating with a Bachelor of Science in Biology. In Newberry, Robin enjoyed some of the best years of her life and made unbreakable friendships, but she looked forward to living again in Clearwater, not only with George, but also near her parents and brother. Back in Florida, Robin worked at a fish hatchery for 7 months and at Pinellas County Utilities for almost 5 years, during which time she married George, became a step-mom to Joshua, and bought two cars and house. All along, Robin knew her education was not complete, so eventually she quit her job and moved to Gainesville to begin graduate school in the Department of Pharmacodynamics, throwing herself, her husband, and her step-son into insurancelessness and disrupting all of their lives. George stayed in Clearwater to be near Joshua, and Robin commuted home on weekends to be 166 with her family. Eventually, George also moved to Gainesville, and he and Robin commuted to Clearwater together to visit the boy on a regular basis. Robin was very lucky to have stumbled her way into Pharmacodynamics, where she learned much and had the opportunity to work with many talented people. As the grand finale to her doctoral academic endeavor draws near, Robin is eager once again to return to life in Clearwater, and yet, more than a little saddened, for, as in Newberry, her times in Gainesville have been unforgettable and her friendships developed there invaluable. She is greatly anticipating the next step in her career, however, and looks forward to a future in science. 167
© Copyright 2026 Paperzz