Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Chem Neuroanat ; 61-62: 239-47, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25172030

ABSTRACT

Reproduction is an energy-expensive process that relies on indicators of energy availability to adjust its proper functioning. The adipokine leptin provides one such metabolic signal, with leptin receptor-expressing neurons at sites widespread within the CNS, including regions associated with the neuroendocrine reproductive axis. One substantial population lies within the hypothalamic ventral premammillary nucleus (PMv), a region itself linked to reproductive control, which may provide a strategic site for the integration of energy availability, sensory and gonadal cues. Here we review our current understanding of leptin and PMv regulation of reproduction, including emerging details about intracellular mechanisms of leptin action at this site.


Subject(s)
Hypothalamus, Posterior/metabolism , Leptin/metabolism , Receptors, Leptin/metabolism , Reproductive Physiological Phenomena , Animals , Humans
2.
Front Neuroanat ; 8: 8, 2014.
Article in English | MEDLINE | ID: mdl-24624061

ABSTRACT

Leptin targets the brain to regulate feeding, neuroendocrine function and metabolism. The leptin receptor is present in hypothalamic centers controlling energy metabolism as well as in the centrally projecting Edinger-Westphal nucleus (EWcp), a region implicated in the stress response and in various aspects of stress-related behaviors. We hypothesized that the stress response by cocaine- and amphetamine-regulated transcript (CART)-producing EWcp-neurons would depend on the animal's energy state. To test this hypothesis, we investigated the effects of changes in energy state (mimicked by low, normal and high leptin levels, which were achieved by 24 h fasting, normal chow and leptin injection, respectively) on the response of CART neurons in the EWcp of rats subjected or not to acute restraint stress. Our data show that leptin treatment alone significantly increases CART mRNA expression in the rat EWcp and that in leptin receptor deficient (db/db) mice, the number of CART producing neurons in this nucleus is reduced. This suggests that leptin has a stimulatory effect on the production of CART in the EWcp under non-stressed condition. Under stressed condition, however, leptin blunts stress-induced activation of EWcp neurons and decreases their CART mRNA expression. Interestingly, fasting, does not influence the stress-induced activation of EWcp-neurons, and specifically EWcp-CART neurons are not activated. These results suggest that the stress response by the EWcp depends to some degree on the animal's energy state, a mechanism that may contribute to a better understanding of the complex interplay between obesity and stress.

3.
Cell Metab ; 15(5): 703-12, 2012 May 02.
Article in English | MEDLINE | ID: mdl-22560222

ABSTRACT

Irs2-mediated insulin/IGF1 signaling in the CNS modulates energy balance and glucose homeostasis; however, the site for Irs2 function is unknown. The hormone leptin mediates energy balance by acting on leptin receptor (LepR-b)-expressing neurons. To determine whether LepR-b neurons mediate the metabolic actions of Irs2 in the brain, we utilized Lepr(cre) together with Irs2(L/L) to ablate Irs2 expression in LepR-b neurons (Lepr(ΔIrs2)). Lepr(ΔIrs2) mice developed obesity, glucose intolerance, and insulin resistance. Leptin action was not altered in young Lepr(ΔIrs2) mice, although insulin-stimulated FoxO1 nuclear exclusion was reduced in Lepr(ΔIrs2) mice. Indeed, deletion of Foxo1 from LepR-b neurons in Lepr(ΔIrs2) mice normalized energy balance, glucose homeostasis, and arcuate nucleus gene expression. Thus, Irs2 signaling in LepR-b neurons plays a crucial role in metabolic sensing and regulation. While not required for leptin action, Irs2 suppresses FoxO1 signaling in LepR-b neurons to promote energy balance and metabolism.


Subject(s)
Forkhead Transcription Factors/metabolism , Insulin Receptor Substrate Proteins/metabolism , Leptin/metabolism , Neurons/metabolism , Receptors, Leptin/metabolism , Animals , Brain/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Energy Metabolism , Female , Forkhead Box Protein O1 , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression , Glucose/genetics , Glucose/metabolism , Glucose Intolerance/genetics , Glucose Intolerance/metabolism , Homeostasis , Insulin/genetics , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Resistance/genetics , Leptin/genetics , Male , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Obesity/genetics , Obesity/metabolism , Receptors, Leptin/genetics , Signal Transduction
4.
Nat Med ; 18(5): 820-3, 2012 May.
Article in English | MEDLINE | ID: mdl-22522563

ABSTRACT

Few effective measures exist to combat the worldwide obesity epidemic(1), and the identification of potential therapeutic targets requires a deeper understanding of the mechanisms that control energy balance. Leptin, an adipocyte-derived hormone that signals the long-term status of bodily energy stores, acts through multiple types of leptin receptor long isoform (LepRb)-expressing neurons (called here LepRb neurons) in the brain to control feeding, energy expenditure and endocrine function(2-4). The modest contributions to energy balance that are attributable to leptin action in many LepRb populations(5-9) suggest that other previously unidentified hypothalamic LepRb neurons have key roles in energy balance. Here we examine the role of LepRb in neuronal nitric oxide synthase (NOS1)-expressing LebRb (LepRb(NOS1)) neurons that comprise approximately 20% of the total hypothalamic LepRb neurons. Nos1(cre)-mediated genetic ablation of LepRb (Lepr(Nos1KO)) in mice produces hyperphagic obesity, decreased energy expenditure and hyperglycemia approaching that seen in whole-body LepRb-null mice. In contrast, the endocrine functions in Lepr(Nos1KO) mice are only modestly affected by the genetic ablation of LepRb in these neurons. Thus, hypothalamic LepRb(NOS1) neurons are a key site of action of the leptin-mediated control of systemic energy balance.


Subject(s)
Energy Metabolism , Hypothalamus/physiology , Leptin/physiology , Neurons/physiology , Nitric Oxide Synthase Type I/physiology , Animals , Mice , Receptors, Leptin/physiology
5.
Cell Metab ; 13(5): 584-91, 2011 May 04.
Article in English | MEDLINE | ID: mdl-21531340

ABSTRACT

Serotonin (5-HT) and leptin play important roles in the modulation of energy balance. Here we investigated mechanisms by which leptin might interact with CNS 5-HT pathways to influence appetite. Although some leptin receptor (LepRb) neurons lie close to 5-HT neurons in the dorsal raphe (DR), 5-HT neurons do not express LepRb. Indeed, while leptin hyperpolarizes some non-5-HT DR neurons, leptin does not alter the activity of DR 5-HT neurons. Furthermore, 5-HT depletion does not impair the anorectic effects of leptin. The serotonin transporter-cre allele (Sert(cre)) is expressed in 5-HT (and developmentally in some non-5-HT) neurons. While Sert(cre) promotes LepRb excision in a few LepRb neurons in the hypothalamus, it is not active in DR LepRb neurons, and neuron-specific Sert(cre)-mediated LepRb inactivation in mice does not alter body weight or adiposity. Thus, leptin does not directly influence 5-HT neurons and does not meaningfully modulate important appetite-related determinants via 5-HT neuron function.


Subject(s)
Appetite , Brain/drug effects , Leptin/pharmacology , Neurons/drug effects , Receptors, Leptin/physiology , Serotonin Plasma Membrane Transport Proteins/physiology , Serotonin/metabolism , Animals , Body Weight/drug effects , Brain/cytology , Brain/metabolism , Electrophysiology , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Immunoenzyme Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways , Neurons/cytology , Neurons/metabolism , Obesity/etiology , Obesity/metabolism
6.
Endocrinology ; 152(3): 979-88, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21209012

ABSTRACT

A recent study systematically characterized the distribution of the long form of the leptin receptor (LepRb) in the mouse brain and showed substantial LepRb mRNA expression in the nonpreganglionic Edinger-Westphal nucleus (npEW) in the rostroventral part of the midbrain. This nucleus hosts the majority of urocortin 1 (Ucn1) neurons in the rodent brain, and because Ucn1 is a potent satiety hormone and electrical lesioning of the npEW strongly decreases food intake, we have hypothesized a role of npEW-Ucn1 neurons in leptin-controlled food intake. Here, we show by immunohistochemistry that npEW-Ucn1 neurons in the mouse contain LepRb and respond to leptin administration with induction of the Janus kinase 2-signal transducer and activator of transcription 3 pathway, both in vivo and in vitro. Furthermore, systemic leptin administration increases the Ucn1 content of the npEW significantly, whereas in mice that lack LepRb (db/db mice), the npEW contains considerably reduced amount of Ucn1. Finally, we reveal by patch clamping of midbrain Ucn1 neurons that leptin administration reduces the electrical firing activity of the Ucn1 neurons. In conclusion, we provide ample evidence for leptin actions that go beyond leptin's well-known targets in the hypothalamus and propose that leptin can directly influence the activity of the midbrain Ucn1 neurons.


Subject(s)
Leptin/metabolism , Neurons/metabolism , Signal Transduction/physiology , Urocortins/metabolism , Animals , Gene Expression , Male , Mesencephalon/cytology , Mice , Patch-Clamp Techniques , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Urocortins/genetics
7.
Brain Res ; 1378: 18-28, 2011 Mar 10.
Article in English | MEDLINE | ID: mdl-21237139

ABSTRACT

Leptin acts via the long form of the leptin receptor (LepRb) on specialized sets of neurons in the brain to modulate diverse functions in concert with energy stores. Previous studies have revealed the distribution of LepRb-expressing neurons in the brain but not the regions to which LepRb neurons project to mediate downstream leptin actions. We utilized LepRb-cre in combination with cre-inducible enhanced green fluorescent protein (EGFP) and farnesylated EGFP (EGFPf) mouse reporter strains to visualize LepRb neurons and their projections, respectively, throughout the brain. The areas containing LepRb soma and projections were relatively circumscribed, as many brain regions contained no detectable EGFP or EGFPf. The highest concentrations of LepRb neurons and LepRb projections were found in the hypothalamus, where the ventral premamillary (PMv), dorsomedial (DMH), and arcuate (ARC) nuclei contained the greatest number of cell bodies, in addition to substantial EGFPf-reactivity. Furthermore, both LepRb soma and projections were present in a few midbrain and brainstem nuclei. Several brain regions including the hypothalamic paraventricular nucleus (PVH), the anteroventral periventricular nucleus (AVPe), and the central nucleus of the amygdala (CeA) contained few LepRb neurons but substantial EGFPf, suggesting that these regions represent targets of LepRb neurons that lie elsewhere in the brain. In some nuclei that contained both soma and projections, the distribution of soma and projections differed, suggesting that these areas transmit leptin-encoded information in a neuroanatomically directional manner.


Subject(s)
Brain Mapping , Brain/cytology , Neural Pathways/cytology , Receptors, Leptin/biosynthesis , Animals , Brain/metabolism , Female , Mice , Neural Pathways/metabolism
8.
J Neurosci ; 30(16): 5713-23, 2010 Apr 21.
Article in English | MEDLINE | ID: mdl-20410123

ABSTRACT

Leptin acts via its receptor (LepRb) to regulate neural circuits in concert with body energy stores. In addition to acting on a number of hypothalamic structures, leptin modulates the mesolimbic dopamine (DA) system. To determine the sites at which LepRb neurons might directly influence the mesolimbic DA system, we examined the distribution of LepRb neurons and their projections within mesolimbic brain regions. Although the ventral tegmental area (VTA) contains DA LepRb neurons, LepRb neurons are absent from the amygdala and striatum. Also, LepRb-EGFPf mice (which label projections from LepRb neurons throughout the brain) reveal that few LepRb neurons project to the nucleus accumbens (NAc). In contrast, the central amygdala (CeA) and its rostral extension receive copious projections from LepRb neurons. Indeed, LepRb-specific anterograde tracing demonstrates (and retrograde tracing confirms) that VTA LepRb neurons project to the extended CeA (extCeA) but not the NAc. Consistently, leptin promotes cAMP response element-binding protein phosphorylation in the extCeA, but not NAc, of leptin-deficient animals. Furthermore, transgenic mice expressing the trans-synaptic tracer wheat germ agglutinin in LepRb neurons reveal the innervation of CeA cocaine- and amphetamine-regulated transcript (CART) neurons by LepRb neurons, and leptin suppresses the increased CeA CART expression of leptin-deficient animals. Thus, LepRb VTA neurons represent a subclass of VTA DA neurons that specifically innervates and controls the extCeA; we hypothesize that these neurons primarily modulate CeA-directed behaviors.


Subject(s)
Amphetamine , Amygdala/physiology , Cocaine , Neurons/physiology , Receptors, Leptin/physiology , Ventral Tegmental Area/physiology , Amphetamine/analysis , Amygdala/chemistry , Animals , Cocaine/analysis , Mice , Mice, Obese , Mice, Transgenic , Neural Pathways/chemistry , Neural Pathways/physiology , Neurons/chemistry , Neurons/classification , Receptors, Leptin/analysis , Transcription, Genetic/physiology , Ventral Tegmental Area/chemistry
9.
Cell Metab ; 10(2): 89-98, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19656487

ABSTRACT

The lateral hypothalamic area (LHA) acts in concert with the ventral tegmental area (VTA) and other components of the mesolimbic dopamine (DA) system to control motivation, including the incentive to feed. The anorexigenic hormone leptin modulates the mesolimbic DA system, although the mechanisms underlying this control have remained incompletely understood. We show that leptin directly regulates a population of leptin receptor (LepRb)-expressing inhibitory neurons in the LHA and that leptin action via these LHA LepRb neurons decreases feeding and body weight. Furthermore, these LHA LepRb neurons innervate the VTA, and leptin action on these neurons restores VTA expression of the rate-limiting enzyme in DA production along with mesolimbic DA content in leptin-deficient animals. Thus, these findings reveal that LHA LepRb neurons link anorexic leptin action to the mesolimbic DA system.


Subject(s)
Dopamine/metabolism , Eating/physiology , Hypothalamic Area, Lateral/metabolism , Leptin/metabolism , Neurons/metabolism , Receptors, Leptin/metabolism , Animals , Body Weight , Gene Knock-In Techniques , Hypothalamic Area, Lateral/cytology , Leptin/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/cytology , Receptors, Leptin/genetics , Ventral Tegmental Area/cytology
10.
Endocrinology ; 150(10): 4541-51, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19628573

ABSTRACT

The medial basal hypothalamus, including the arcuate nucleus (ARC) and the ventromedial hypothalamic nucleus (VMH), integrates signals of energy status to modulate metabolism and energy balance. Leptin and feeding regulate the mammalian target of rapamycin complex 1 (mTORC1) in the hypothalamus, and hypothalamic mTORC1 contributes to the control of feeding and energy balance. To determine the mechanisms by which leptin modulates mTORC1 in specific hypothalamic neurons, we immunohistochemically assessed the mTORC1-dependent phosphorylation of ribosomal protein S6 (pS6). In addition to confirming the modulation of ARC mTORC1 activity by acute leptin treatment, this analysis revealed the robust activation of mTORC1-dependent ARC pS6 in response to fasting and leptin deficiency in leptin receptor-expressing Agouti-related protein neurons. In contrast, fasting and leptin deficiency suppress VMH mTORC1 signaling. The appropriate regulation of ARC mTORC1 by mutant leptin receptor isoforms correlated with their ability to suppress the activity of Agouti-related protein neurons, suggesting the potential stimulation of mTORC1 by the neuronal activity. Indeed, fasting- and leptin deficiency-induced pS6-immunoreactivity (IR) extensively colocalized with c-Fos-IR in ARC and VMH neurons. Furthermore, ghrelin, which activates orexigenic ARC neurons, increased ARC mTORC1 activity and induced colocalized pS6- and c-Fos-IR. Thus, neuronal activity promotes mTORC1/pS6 in response to signals of energy deficit. In contrast, insulin, which activates mTORC1 via the phosphatidylinositol 3-kinase pathway, increased ARC and VMH pS6-IR in the absence of neuronal activation. The regulation of mTORC1 in the basomedial hypothalamus thus varies by cell and stimulus type, as opposed to responding in a uniform manner to nutritional and hormonal perturbations.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Leptin/metabolism , Nutritional Status , Receptors, Leptin/metabolism , Transcription Factors/metabolism , Agouti-Related Protein/metabolism , Animals , Energy Metabolism , Fasting/physiology , Insulin/metabolism , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Multiprotein Complexes , Neurons/metabolism , Phosphorylation , Proteins , Ribosomal Protein S6/metabolism , Ribosomal Protein S6 Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases , Ventromedial Hypothalamic Nucleus/physiology
11.
J Neurosci ; 29(10): 3138-47, 2009 Mar 11.
Article in English | MEDLINE | ID: mdl-19279251

ABSTRACT

Leptin acts via its receptor (LepRb) on specific CNS neurons to signal the adequacy of long-term energy stores, thereby permitting the expenditure of resources on energy-intensive processes such as reproduction. The ventral premammillary nucleus of the hypothalamus (PMv), which has been implicated in the stimulation of gonadotropin release by olfactory cues, contains numerous LepRb neurons, suggesting a potential role for LepRb PMv neurons in transmitting both metabolic and odorant signals to the neuroendocrine reproductive system. Indeed, Fos immunoreactivity and electrophysiologic recordings revealed the direct activation of LepRb PMv neurons by leptin, and exposure to odors from mice of the opposite sex promoted Fos immunoreactivity (Fos-IR) in many LepRb PMv neurons. To determine the regions innervated by the LepRb PMv neurons, we used two novel cre-activated tract-tracing systems in Lepr(cre) animals; data from these systems and from standard tracing techniques revealed that LepRb PMv neurons project to a subset of the regions, including the preoptic area, that are innervated by the PMv as a whole. Furthermore, the retrograde accumulation in LepRb PMv neurons of a trans-synaptic tracer from GnRH neurons revealed the direct innervation of GnRH neurons by many LepRb PMv neurons. Thus, LepRb PMv neurons sense metabolic and sexual odorant cues and project to the rostral hypothalamus to directly innervate GnRH neurons. These results are consistent with a role for LepRb PMv neurons in regulating the reproductive axis in response to metabolic and odorant stimuli.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/chemistry , Neurons/metabolism , Receptors, Leptin/physiology , Receptors, Odorant/analysis , Sex Attractants/administration & dosage , Animals , Female , Gene Knock-In Techniques , Gonadotropin-Releasing Hormone/analysis , Hypothalamus/drug effects , Hypothalamus/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Nerve Net/chemistry , Nerve Net/drug effects , Nerve Net/metabolism , Neurons/chemistry , Neurons/drug effects , Receptors, Leptin/genetics , Receptors, Odorant/physiology , Sex Attractants/physiology
12.
Diabetes ; 58(5): 1192-200, 2009 May.
Article in English | MEDLINE | ID: mdl-19208909

ABSTRACT

OBJECTIVE: To characterize the hormonal milieu and adipose gene expression in response to catch-up growth (CUG), a growth pattern associated with obesity and diabetes risk, in a mouse model of low birth weight (LBW). RESEARCH DESIGN AND METHODS: ICR mice were food restricted by 50% from gestational days 12.5-18.5, reducing offspring birth weight by 25%. During the suckling period, dams were either fed ad libitum, permitting CUG in offspring, or food restricted, preventing CUG. Offspring were killed at age 3 weeks, and gonadal fat was removed for RNA extraction, array analysis, RT-PCR, and evaluation of cell size and number. Serum insulin, thyroxine (T4), corticosterone, and adipokines were measured. RESULTS: At age 3 weeks, LBW mice with CUG (designated U-C) had body weight comparable with controls (designated C-C); weight was reduced by 49% in LBW mice without CUG (designated U-U). Adiposity was altered by postnatal nutrition, with gonadal fat increased by 50% in U-C and decreased by 58% in U-U mice (P < 0.05 vs. C-C mice). Adipose expression of the lipogenic genes Fasn, AccI, Lpin1, and Srebf1 was significantly increased in U-C compared with both C-C and U-U mice (P < 0.05). Mitochondrial DNA copy number was reduced by >50% in U-C versus U-U mice (P = 0.014). Although cell numbers did not differ, mean adipocyte diameter was increased in U-C and reduced in U-U mice (P < 0.01). CONCLUSIONS: CUG results in increased adipose tissue lipogenic gene expression and adipocyte diameter but not increased cellularity, suggesting that catch-up fat is primarily associated with lipogenesis rather than adipogenesis in this murine model.


Subject(s)
Adipocytes/cytology , Gene Expression Regulation, Developmental , Growth/physiology , Adipose Tissue/anatomy & histology , Adipose Tissue/growth & development , Animals , Birth Weight , Cell Size , Female , Glucose/metabolism , Hyperphagia/epidemiology , Male , Mice , Mice, Inbred ICR , Midline Thalamic Nuclei/anatomy & histology , Midline Thalamic Nuclei/growth & development , Pregnancy
13.
Cell Metab ; 9(2): 117-23, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19187770

ABSTRACT

Leptin signals the repletion of fat stores, acting in the CNS to permit energy utilization by a host of autonomic and neuroendocrine processes and to decrease feeding. While much recent research has focused on the leptin-regulated circuitry of the hypothalamic arcuate nucleus (ARC), the majority of brain leptin receptor (LepRb)-expressing neurons lie outside the ARC in other CNS regions known to modulate energy balance. Each set of LepRb neurons throughout the brain presumably mediates unique aspects of leptin action, and understanding the function for LepRb-expressing neurons throughout the brain represents a crucial next step in the study of energy homeostasis.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Brain/metabolism , Leptin/metabolism , Receptors, Leptin/metabolism , Central Nervous System , Energy Metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism
14.
Transgenic Res ; 16(5): 665-70, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17570071

ABSTRACT

The identification of correctly targeted embryonic stem (ES) cell clones from among the large number of random integrants that result from most selection paradigms remains an important hurdle in the generation of animals bearing homologously targeted transgenes. Given the limitations inherent to Southern blotting and standard PCR, we utilized quantitative real-time polymerase chain reaction (qPCR) to rapidly identify murine ES cell clones containing insertions at the correct genomic locus. Importantly, this approach is useful for screening ES clones from conditional/insertional "knock-in" strategies in which there is no loss of genetic material. Simple validation avoids the generation of assays prone to false negative results. In this method, probe and primer sets that span an insertion site detect and quantify the unperturbed gene relative to an irrelevant reference gene, allowing ES cell clones to be screened for loss of detection of one copy of the gene (functional loss of homozygousity (LOH)) that occurs when the normal DNA is disrupted by the insertion event. Simply stated, detected gene copy number falls from two to one in correctly targeted clones. We have utilized such easily designed and validated qPCR LOH assays to rapidly and accurately identify insertions in multiple target sites (including the Lepr and mTOR loci) in murine ES cells, in order to generate transgenic animals.


Subject(s)
Embryonic Stem Cells/cytology , Polymerase Chain Reaction/methods , Animals , Base Sequence , Blotting, Southern , DNA/chemistry , DNA Primers/chemistry , Genetic Techniques , Genetic Vectors , Genome , Loss of Heterozygosity , Mice , Models, Genetic , Molecular Sequence Data
15.
J Clin Invest ; 117(5): 1354-60, 2007 May.
Article in English | MEDLINE | ID: mdl-17415414

ABSTRACT

The adipose-derived hormone, leptin, acts via its receptor (LRb) to convey the status of body energy stores to the brain, decreasing feeding and potentiating neuroendocrine energy expenditure. The failure of high levels of leptin in most obese individuals to promote weight loss defines a state of diminished responsiveness to increased leptin, termed leptin resistance. Leptin stimulates the phosphorylation of several tyrosine residues on LRb to mediate leptin action. We homologously replaced LRb in mice with a receptor with a mutation in one of these sites (Tyr985) in order to examine its role in leptin action and signal attenuation in vivo. Mice homozygous for this mutation are neuroendocrinologically normal, but females demonstrate decreased feeding, decreased expression of orexigenic neuropeptides, protection from high-fat diet-induced obesity, and increased leptin sensitivity in a sex-biased manner. Thus, leptin activates autoinhibitory signals via LRb Tyr985 to attenuate the anti-adiposity effects of leptin, especially in females, potentially contributing to leptin insensitivity in obesity.


Subject(s)
Endocrine System/physiology , Receptors, Cell Surface/deficiency , Receptors, Cell Surface/physiology , Signal Transduction/genetics , Thinness/genetics , Thinness/metabolism , Amino Acid Substitution/genetics , Animals , Female , Leptin/antagonists & inhibitors , Leptin/physiology , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Receptors, Leptin , Sensitivity and Specificity , Sex Factors , Thinness/physiopathology , Tyrosine/genetics
16.
Obesity (Silver Spring) ; 14 Suppl 5: 208S-212S, 2006 Aug.
Article in English | MEDLINE | ID: mdl-17021368

ABSTRACT

The increasing incidence of obesity in developed nations represents an ever-growing challenge to health care by promoting diabetes and other diseases. The discovery of the hormone, leptin, a decade ago has facilitated the acquisition of new knowledge regarding the regulation of energy balance. A great deal remains to be discovered regarding the molecular and anatomic actions of leptin, however. Here, we discuss the mechanisms by which leptin activates intracellular signals, the roles that these signals play in leptin action in vivo, and sites of leptin action in vivo. Using "reporter" mice, in which LRb-expressing (long form of the leptin receptor) neurons express the histological marker, beta-galactosidase, coupled with the detection of LRb-mediated signal transducer and activator of transcription 3 signaling events, we identified LRb expression in neuronal populations both within and outside the hypothalamus. Understanding the regulation and physiological function of these myriad sites of central leptin action will be a crucial next step in the quest to understand mechanisms of leptin action and energy balance.


Subject(s)
Central Nervous System/physiology , Energy Metabolism/physiology , Leptin/physiology , Obesity/etiology , Receptors, Cell Surface/metabolism , Signal Transduction , Animals , Gene Expression , Homeostasis/physiology , Humans , Mice , Mice, Transgenic , Multienzyme Complexes , Neurons/physiology , Obesity/metabolism , Receptors, Leptin
18.
J Neurosci ; 25(5): 1240-8, 2005 Feb 02.
Article in English | MEDLINE | ID: mdl-15689562

ABSTRACT

Insulin receptor substrates (Irs-proteins) integrate signals from the insulin and insulin-like growth factor-1 (IGF1) receptors with other processes to control cellular growth, function, and survival. Here, we show that Irs2 promoted the maturation and survival of photoreceptors in the murine retina immediately after birth. Irs2 was mainly localized to the outer plexiform layer as well as to photoreceptor inner segments. It was also seen in ganglion cells and inner plexiform layer but in smaller amounts. Compared with control littermates, Irs2 knock-out mice lose 10% of their photoreceptors 1 week after birth and up to 50% by 2 weeks of age as a result of increased apoptosis. The surviving photoreceptor cells developed short organized segments, which displayed proportionally diminished but otherwise normal electrical function. However, IGF1-stimulated Akt phosphorylation was barely detected, and cleaved/activated caspase-3 was significantly elevated in isolated retinas of Irs2-/- mice. When diabetes was prevented, which allowed the Irs2-/- mice to survive for 2 years, most photoreceptor cells were lost by 16 months of age. Because apoptosis is the final common pathway in photoreceptor degeneration, pharmacological strategies that increase Irs2 expression or function in photoreceptor cells could be a general treatment for blinding diseases such as retinitis pigmentosa.


Subject(s)
Eye Proteins/physiology , Phosphoproteins/physiology , Photoreceptor Cells/cytology , Age Factors , Animals , Animals, Newborn , Apoptosis , Cell Survival , Diabetic Retinopathy/genetics , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/physiopathology , Eye Proteins/genetics , Gene Deletion , Homeodomain Proteins/genetics , Homeodomain Proteins/physiology , Hyperglycemia/genetics , Hyperinsulinism/genetics , Insulin Receptor Substrate Proteins , Insulin Resistance , Insulin-Like Growth Factor I/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Knockout , Phosphoproteins/deficiency , Phosphoproteins/genetics , Phosphorylation , Photic Stimulation , Photoreceptor Cells/metabolism , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Retinal Ganglion Cells/metabolism , Signal Transduction , Trans-Activators/genetics , Trans-Activators/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...