Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 201
Filter
1.
Nat Commun ; 4: 2049, 2013.
Article in English | MEDLINE | ID: mdl-23804023

ABSTRACT

Emerging evidence suggests that new cells, including neurons, can be generated within the adult hypothalamus, suggesting the existence of a local neural stem/progenitor cell niche. Here, we identify α-tanycytes as key components of a hypothalamic niche in the adult mouse. Long-term lineage tracing in vivo using a GLAST::CreER(T2) conditional driver indicates that α-tanycytes are self-renewing cells that constitutively give rise to new tanycytes, astrocytes and sparse numbers of neurons. In vitro studies demonstrate that α-tanycytes, but not ß-tanycytes or parenchymal cells, are neurospherogenic. Distinct subpopulations of α-tanycytes exist, amongst which only GFAP-positive dorsal α2-tanycytes possess stem-like neurospherogenic activity. Fgf-10 and Fgf-18 are expressed specifically within ventral tanycyte subpopulations; α-tanycytes require fibroblast growth factor signalling to maintain their proliferation ex vivo and elevated fibroblast growth factor levels lead to enhanced proliferation of α-tanycytes in vivo. Our results suggest that α-tanycytes form the critical component of a hypothalamic stem cell niche, and that local fibroblast growth factor signalling governs their proliferation.


Subject(s)
Aging/metabolism , Ependymoglial Cells/metabolism , Fibroblast Growth Factor 10/metabolism , Fibroblast Growth Factors/metabolism , Hypothalamus/cytology , Neural Stem Cells/metabolism , Third Ventricle/cytology , Animals , Cell Proliferation/drug effects , Ependymoglial Cells/cytology , Ependymoglial Cells/drug effects , Epidermal Growth Factor/pharmacology , Excitatory Amino Acid Transporter 1/metabolism , Glial Fibrillary Acidic Protein/metabolism , Immunohistochemistry , Integrases/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neuroglia/cytology , Neuroglia/drug effects , Neuroglia/metabolism , Signal Transduction/drug effects , Spheroids, Cellular/cytology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
2.
Science ; 294(5549): 2166-70, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11739957

ABSTRACT

The adverse metabolic consequences of obesity are best predicted by the quantity of visceral fat. Excess glucocorticoids produce visceral obesity and diabetes, but circulating glucocorticoid levels are normal in typical obesity. Glucocorticoids can be produced locally from inactive 11-keto forms through the enzyme 11beta hydroxysteroid dehydrogenase type 1 (11beta HSD-1). We created transgenic mice overexpressing 11beta HSD-1 selectively in adipose tissue to an extent similar to that found in adipose tissue from obese humans. These mice had increased adipose levels of corticosterone and developed visceral obesity that was exaggerated by a high-fat diet. The mice also exhibited pronounced insulin-resistant diabetes, hyperlipidemia, and, surprisingly, hyperphagia despite hyperleptinemia. Increased adipocyte 11beta HSD-1 activity may be a common molecular etiology for visceral obesity and the metabolic syndrome.


Subject(s)
Adipose Tissue/enzymology , Disease Models, Animal , Hydroxysteroid Dehydrogenases/metabolism , Metabolic Syndrome , Obesity/enzymology , Obesity/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1 , Abdomen , Adipocytes/cytology , Adipocytes/pathology , Adipose Tissue/metabolism , Animals , Body Composition , Cell Size , Corticosterone/blood , Corticosterone/metabolism , Dietary Fats/administration & dosage , Eating , Gene Targeting , Humans , Hydroxysteroid Dehydrogenases/genetics , Hyperglycemia/metabolism , Hyperinsulinism/metabolism , Insulin Resistance , Leptin/metabolism , Lipid Metabolism , Lipids/blood , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Transgenic , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Viscera , Weight Gain
3.
J Clin Endocrinol Metab ; 86(7): 3284-91, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11443202

ABSTRACT

Leptin signals the status of energy reserves to the brain. Leptin stimulates biosynthesis of TRH in vitro and influences the activity of the hypothalamic-pituitary-thyroid axis in vivo in rodents. Because blood levels of both leptin and TSH display diurnal variation with a distinct nocturnal rise, we sought to determine whether a relationship exists between fluctuations in circulating leptin and TSH. We measured serum leptin and TSH levels every 7 min for 24 h in five healthy men and found that both leptin and TSH levels are highly organized and pulsatile. A similar pattern of leptin and TSH rhythms was observed, with TSH and leptin levels reaching a nadir in late morning and a peak in the early morning hours. Importantly, cosinor analysis on the absolute leptin and TSH levels revealed a statistically significant fit for a 24-h period and the two hormones showed similar probabilities of rhythm and superimposable peak values. Furthermore, this study shows a strong positive Pearson correlation between the 24-h patterns of variability of leptin and TSH in healthy subjects. Finally, the ultradian fluctuations in leptin levels showed pattern synchrony with those of TSH as determined by cross-correlation analysis, by cross-approximate enthropy and Bayessian analysis applied independently. To further explore whether these associations could reflect an underlying regulation of TSH secretion by leptin, we also studied frequently sampled leptin and TSH levels in four brothers, members of a family with leptin deficiency (one normal homozygote, two heterozygotes, and one leptin-deficient homozygote). Leptin levels of the homozygous leptin-deficient subject are detectable but bioinactive, and the rhythm of his TSH is disorganized. 24-h pattern of leptin and TSH variability in the heterozygous subjects, although significantly correlated, showed a weaker correlation compared with the strong correlation in the normal subjects. These data are consistent with the possibility that leptin may regulate TSH pulsatility and circadian rhythmicity, but interventional studies are needed to definitively prove whether leptin regulates the minute-to-minute oscillations and ultradian rhythm of TSH levels.


Subject(s)
Leptin/deficiency , Leptin/metabolism , Periodicity , Thyrotropin/metabolism , Adult , Circadian Rhythm , Heterozygote , Homozygote , Humans , Leptin/analysis , Leptin/genetics , Male , Thyrotropin/blood , Thyrotropin-Releasing Hormone , Thyroxine/blood , Triiodothyronine/blood
7.
J Clin Invest ; 107(3): 379-86, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11160162

ABSTRACT

Several lines of investigation suggest that the hypothalamic neuropeptide melanin-concentrating hormone (MCH) regulates body weight in mammals. Obese mice lacking functional leptin overexpress the MCH message in the fed or fasted state. Acute intracerebroventricular injection of MCH increases energy intake in rats. Mice lacking the MCH gene are lean. To test the hypothesis that chronic overexpression of MCH in mice causes obesity, we produced transgenic mice that overexpress MCH (MCH-OE) in the lateral hypothalamus at approximately twofold higher levels than normal mice. On the FVB genetic background, homozygous transgenic animals fed a high-fat diet ate 10% more and were 12% heavier at 13 weeks of age than wild-type animals, and they had higher systemic leptin levels. Blood glucose levels were higher both preprandially and after an intraperitoneal glucose injection. MCH-OE animals were insulin-resistant, as demonstrated by markedly higher plasma insulin levels and a blunted response to insulin; MCH-OE animals had only a 5% decrease in blood glucose after insulin administration, compared with a 31% decrease in wild-type animals. MCH-OE animals also exhibited a twofold increase in islet size. To evaluate the contribution of genetic background to the predisposition to obesity seen in MCH-OE mice, the transgene was bred onto the C57BL/6J background. Heterozygote C57BL/6J mice expressing the transgene showed increased body weight on a standard diet, confirming that MCH overexpression can lead to obesity.


Subject(s)
Hypothalamic Hormones/genetics , Hypothalamus/metabolism , Insulin Resistance , Melanins/genetics , Obesity/genetics , Pituitary Hormones/genetics , Adipose Tissue/metabolism , Animals , Blood Glucose/analysis , Body Weight , Eating , Glucose Tolerance Test , Homeostasis , Hypothalamic Hormones/biosynthesis , Leptin/blood , Male , Melanins/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/metabolism , Pituitary Hormones/biosynthesis , Time Factors
8.
J Clin Invest ; 107(1): 111-20, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11134186

ABSTRACT

Starvation causes a rapid reduction in thyroid hormone levels in rodents. This adaptive response is caused by a reduction in thyrotropin-releasing hormone (TRH) expression that can be reversed by the administration of leptin. Here we examined hypothalamic signaling pathways engaged by leptin to upregulate TRH gene expression. As assessed by leptin-induced expression of suppressor of cytokine signaling-3 (SOCS-3) in fasted rats, TRH neurons in the paraventricular nucleus are activated directly by leptin. To a greater degree, they also contain melanocortin-4 receptors (MC4Rs), implying that leptin can act directly or indirectly by increasing the production of the MC4R ligand, alpha-melanocyte stimulating hormone (alpha-MSH), to regulate TRH expression. We further demonstrate that both pathways converge on the TRH promoter. The melanocortin system activates the TRH promoter through the phosphorylation and DNA binding of the cAMP response element binding protein (CREB), and leptin signaling directly regulates the TRH promoter through the phosphorylation of signal transducer and activator of transcription 3 (Stat3). Indeed, a novel Stat-response element in the TRH promoter is necessary for leptin's effect. Thus, the TRH promoter is an ideal target for further characterizing the integration of transcriptional pathways through which leptin acts.


Subject(s)
Leptin/pharmacology , Receptors, Peptide/metabolism , Repressor Proteins , Thyrotropin-Releasing Hormone/genetics , Transcription Factors , Animals , Base Sequence , Binding Sites/genetics , DNA/genetics , Fasting/metabolism , Gene Expression Regulation/drug effects , Humans , Male , Models, Biological , Molecular Sequence Data , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Promoter Regions, Genetic , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 4 , Receptors, Leptin , Receptors, Peptide/genetics , Signal Transduction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , alpha-MSH/metabolism , alpha-MSH/pharmacology
9.
J Biol Chem ; 276(7): 4747-55, 2001 Feb 16.
Article in English | MEDLINE | ID: mdl-11085989

ABSTRACT

The protein tyrosine phosphatase SHP-2 has been proposed to serve as a regulator of leptin signaling, but its specific roles are not fully examined. To directly investigate the role of SHP-2, we employed dominant negative strategies in transfected cells. We show that a catalytically inactive mutant of SHP-2 blocks leptin-stimulated ERK phosphorylation by the long leptin receptor, ObRb. SHP-2, lacking two C-terminal tyrosine residues, partially inhibits ERK phosphorylation. We find similar effects of the SHP-2 mutants after examining stimulation of an ERK-dependent egr-1 promoter-construct by leptin. We also demonstrate ERK phosphorylation and egr-1 mRNA expression in the hypothalamus by leptin. Analysis of signaling by ObRb lacking intracellular tyrosine residues or by the short leptin receptor, ObRa, enabled us to conclude that two pathways are critical for ERK activation. One pathway does not require the intracellular domain of ObRb, whereas the other pathway requires tyrosine residue 985 of ObRb. The phosphatase activity of SHP-2 is required for both pathways, whereas activation of ERK via Tyr-985 of ObRb also requires tyrosine phosphorylation of SHP-2. SHP-2 is thus a positive regulator of ERK by leptin receptors, and both the adaptor function and the phosphatase activity of SHP-2 are critical for this regulation.


Subject(s)
Carrier Proteins/metabolism , Immediate-Early Proteins , MAP Kinase Signaling System , Mitogen-Activated Protein Kinases/metabolism , Protein Tyrosine Phosphatases/physiology , Proto-Oncogene Proteins , Receptors, Cell Surface , Animals , CHO Cells , Carrier Proteins/chemistry , Cricetinae , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Early Growth Response Protein 1 , Hypothalamus/metabolism , Intracellular Signaling Peptides and Proteins , Janus Kinase 2 , Leptin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 3 , Models, Biological , Mutation , Phosphorylation , Promoter Regions, Genetic , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protein Tyrosine Phosphatases/genetics , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/physiology , RNA, Messenger/biosynthesis , Receptors, Leptin , STAT3 Transcription Factor , Trans-Activators/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription, Genetic , Transfection
11.
J Biol Chem ; 275(51): 40649-57, 2000 Dec 22.
Article in English | MEDLINE | ID: mdl-11018044

ABSTRACT

During leptin signaling, each of the phosphorylated tyrosine residues on the long form of the leptin receptor (LRb) mediates distinct signals. Phosphorylated Tyr(1138) binds STAT3 to mediate its tyrosine phosphorylation and transcriptional activation, while phosphorylated Tyr(985) binds the tyrosine phosphatase SHP-2 and reportedly mediates both activation of ERK kinases and inhibition of LRb-mediated STAT3 activation. We show here that although mutation of Tyr(985) does not alter STAT3 signaling by erythropoietin receptor-LRb (ELR) chimeras in transfected 293 cells at short times of stimulation, this mutation enhances STAT3 signaling at longer times of stimulation (>6 h). These data suggest that Tyr(985) may mediate feedback inhibition of LRb signaling by an LRb-induced LRb inhibitor, such as SOCS3. Indeed, SOCS3 binds specifically to phosphorylated Tyr(985) of LRb, and SOCS3 fails to inhibit transcription by ELR following mutation of Tyr(985), suggesting that SOCS3 inhibits LRb signaling by binding to phosphorylated Tyr(985). Additionally, overexpression of SOCS3, but not SHP-2, impairs ELR signaling, and the overexpression of SHP-2 blunts SOCS3-mediated inhibition of ELR signaling. Thus, our data suggest that in addition to mediating SHP-2 binding and ERK activation during acute stimulation, Tyr(985) of LRb mediates feedback inhibition of LRb signaling by binding to LRb-induced SOCS3.


Subject(s)
Carrier Proteins/antagonists & inhibitors , Feedback , Proteins/physiology , Receptors, Cell Surface , Repressor Proteins , Signal Transduction , Transcription Factors , Tyrosine/metabolism , Carrier Proteins/chemistry , Cell Line , Humans , Receptors, Leptin , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
12.
Am J Physiol Endocrinol Metab ; 279(4): E838-45, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11001766

ABSTRACT

Exposure to high-fat diets for prolonged periods results in positive energy balance and obesity, but little is known about the initial physiological and neuroendocrine response of obesity-susceptible strains to high-fat feeding. To assess responses of C57BL/6J mice to high- and low-fat diets, we quantitated the hypothalamic expression of neuropeptides implicated in weight regulation and neuroendocrine function over a 2-wk period. Exposure to high-fat diet increased food consumption over a 2-day period during which leptin levels were increased when assessed by a frequent sampling protocol [area under the curve (AUC): 134.6 +/- 10.3 vs. 100 +/- 12.3, P = 0.03 during first day and 126.5 +/- 8.2 vs. 100 +/- 5.2, P = 0.02 during second day]. During this period, hypothalamic expression of neuropeptide Y (NPY) and agouti-related protein (AgRP) decreased by approximately 30 and 50%, respectively (P < 0.001). After 1 wk, both caloric intake and hypothalamic expression of NPY and AgRP returned toward baseline. After 2 wk, cumulative caloric intake was again higher in the high-fat group, and now proopiomelanocortin (POMC) was elevated by 76% (P = 0.01). This study demonstrates that high-fat feeding induces hyperphagia, hyperleptinemia, and transient suppression of orexigenic neuropeptides during the first 2 days of diet. The subsequent induction of POMC may be a second defense against obesity. Attempts to understand the hypothalamic response to high-fat feeding must examine the changes as they develop over time.


Subject(s)
Dietary Fats/pharmacology , Hypothalamus/metabolism , Neuropeptides/biosynthesis , Obesity/metabolism , Repressor Proteins , Transcription Factors , Agouti-Related Protein , Animals , Area Under Curve , Body Composition/drug effects , Body Weight/drug effects , Corticosterone/blood , Eating/drug effects , Energy Intake/drug effects , Fatty Acids, Nonesterified/blood , Insulin/blood , Intercellular Signaling Peptides and Proteins , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Neuropeptide Y/biosynthesis , Neuropeptide Y/genetics , Neuropeptides/genetics , Obesity/etiology , Pro-Opiomelanocortin/biosynthesis , Pro-Opiomelanocortin/genetics , Protein Biosynthesis , Proteins/genetics , RNA, Messenger/biosynthesis , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins
13.
Trends Endocrinol Metab ; 11(8): 327-32, 2000 Oct.
Article in English | MEDLINE | ID: mdl-10996528

ABSTRACT

The discovery of leptin in the mid-1990s has focused attention on the role of proteins secreted by adipose tissue. Leptin has profound effects on appetite and energy balance, and is also involved in the regulation of neuroendocrine and immune function. Sex steroid and glucocorticoid metabolism in adipose tissue has been implicated as a determinant of body fat distribution and cardiovascular risk. Other adipose products, for example, proinflammatory cytokines, complement factors and components of the coagulation/fibrinolytic cascade, may mediate the metabolic and cardiovascular complications associated with obesity.


Subject(s)
Adipose Tissue/physiology , Endocrine Glands/physiology , Hormones/physiology , Adipose Tissue/metabolism , Animals , Hormones/biosynthesis , Humans
14.
J Biol Chem ; 275(46): 36124-33, 2000 Nov 17.
Article in English | MEDLINE | ID: mdl-10967095

ABSTRACT

The hypothalamic-pituitary-thyroid axis is down-regulated during starvation, and falling levels of leptin are a critical signal for this adaptation, acting to suppress preprothyrotropin-releasing hormone (prepro-TRH) mRNA expression in the paraventricular nucleus of the hypothalamus. This study addresses the mechanism for this regulation, using primary cultures of fetal rat hypothalamic neurons as a model system. Leptin dose-dependently stimulated a 10-fold increase in pro-TRH biosynthesis, with a maximum response at 10 nm. TRH release was quantified using immunoprecipitation, followed by isoelectric focusing gel electrophoresis and specific TRH radioimmunoassay. Leptin stimulated TRH release by 7-fold. Immunocytochemistry revealed that a substantial population of cells expressed TRH or leptin receptors and that 8-13% of those expressing leptin receptors coexpressed TRH. Leptin produced a 5-fold induction of luciferase activity in CV-1 cells transfected with a TRH promoter and the long form of the leptin receptor cDNA. Although the above data are consistent with a direct ability of leptin to promote TRH biosynthesis through actions on TRH neurons, addition of alpha-melanocyte-stimulating hormone produced a 3.5-fold increase in TRH biosynthesis and release, whereas neuropeptide Y treatment suppressed pro-TRH biosynthesis approximately 3-fold. Furthermore, the melanocortin-4 receptor antagonist SHU9119 partially inhibited leptin-stimulated TRH release from the neuronal culture. Consequently, our data suggest that leptin regulates the TRH neurons through both direct and indirect pathways.


Subject(s)
Leptin/metabolism , Protein Precursors/biosynthesis , Receptors, Cell Surface , Thyrotropin-Releasing Hormone/biosynthesis , Animals , Antibody Specificity , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Fluorescent Antibody Technique , Hypothalamus/cytology , Hypothalamus/embryology , Models, Biological , Neurons/drug effects , Neurons/metabolism , Neuropeptide Y/pharmacology , Promoter Regions, Genetic , Protein Precursors/genetics , Pyrrolidonecarboxylic Acid/analogs & derivatives , RNA, Messenger/genetics , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Sprague-Dawley , Receptors, Leptin , Signal Transduction/drug effects , Thyrotropin-Releasing Hormone/genetics , Thyrotropin-Releasing Hormone/metabolism , alpha-MSH/pharmacology
16.
Front Neuroendocrinol ; 21(3): 263-307, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10882542

ABSTRACT

The discovery of leptin has enhanced understanding of the interrelationship between adipose energy stores and neuronal circuits in the brain involved in energy balance and regulation of the neuroendocrine axis. Leptin levels are dependent on the status of fat stores as well as changes in energy balance as a result of fasting and overfeeding. Although leptin was initially thought to serve mainly as an anti-satiety hormone, recent studies have shown that it mediates the adaptation to fasting. Furthermore, leptin has been implicated in the regulation of the reproductive, thyroid, growth hormone, and adrenal axes, independent of its role in energy balance. Although it is widely known that leptin acts on hypothalamic neuronal targets to regulate energy balance and neuroendocrine function, the specific neuronal populations mediating leptin action on feeding behavior and autonomic and neuroendocrine function are not well understood. In this review, we have discussed how leptin engages arcuate hypothalamic neurons expressing putative orexigenic peptides, e.g., neuropeptide Y and agouti-regulated peptide, and anorexigenic peptides, e.g., pro-opiomelanocortin (precursor of alpha-melanocyte-stimulating hormone) and cocaine- and amphetamine-regulated transcript. We show that leptin's effects on energy balance and the neuroendocrine axis are mediated by projections to other hypothalamic nuclei, e.g., paraventricular, lateral, and perifornical areas, as well as other sites in the brainstem, spinal cord, and cortical and subcortical regions.


Subject(s)
Leptin/physiology , Neurosecretory Systems/physiology , Animals , Biological Transport , Brain/metabolism , Energy Metabolism/physiology , Homeostasis/physiology , Humans , Leptin/metabolism
17.
Endocrinology ; 141(6): 1955-61, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10830277

ABSTRACT

Leptin is an adipocyte-derived hormone that acts in specific regions of the brain to regulate body weight and neuroendocrine function. The mechanism by which leptin enters the brain is unknown. We previously reported that rat brain microvessels, which constitute the blood-brain barrier, contain large amounts of messenger RNA encoding a short form of the leptin receptor (ObRa), suggesting that this site may be important for receptor-mediated transport of leptin into the brain. The purpose of this study was to determine whether ObRa is capable of transcellular transport of intact leptin. A transwell system in which Madin-Darby Canine Kidney (MDCK) cells stably expressing ObRa are grown in a monolayer was used to determine receptor distribution on apical or basolateral cell surfaces and the capacity for directional transport of 125I-leptin. Binding of 125I-leptin was greater on the apical vs. the basolateral cell surface and transport of 125I-leptin occurred only in the apical to basolateral direction. 11% of transported radioactivity appearing in the basolateral chamber represented intact leptin as assessed by TCA precipitation analysis and by SDS-PAGE. Parental MDCK cells did not express leptin receptors and did not bind or transport 125I-leptin. Epidermal growth factor (EGF) binding and transport via endogenous EGF receptors in MDCK cells also was assessed. In contrast to leptin, specific binding of 125I-EGF occurred primarily on the basolateral cell surface and transport of 125I-EGF occurred predominantly in the basolateral to apical direction. These data show that ObRa is preferentially targeted to the apical cell membrane in MDCK cells and that leptin transport occurs, albeit at a low rate, in a unidirectional manner in the apical to basolateral direction. These findings may be relevant to the putative role of ObRa in receptor-mediated transport of leptin from the circulation into the brain.


Subject(s)
Carrier Proteins/metabolism , Leptin/metabolism , Receptors, Cell Surface , Animals , Biological Transport , Carrier Proteins/genetics , Cell Line , Cell Membrane/metabolism , Dogs , Epidermal Growth Factor/metabolism , Gene Expression , Iodine Radioisotopes , Kidney , Receptors, Leptin , Transfection
18.
Endocrinology ; 141(7): 2328-39, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10875232

ABSTRACT

To determine whether leptin signal transduction is exerted directly upon insulin-sensitive tissues in vivo, we examined the ability of iv leptin to acutely stimulate phosphorylation of STAT3, STAT1, and MAPK, and activities of PI 3-kinase and Akt, in insulin-sensitive tissues of normal rats. Both leptin (1 mg/kg iv x 3 min) and insulin (10 U/kg iv x 3 min) stimulated tyrosine phosphorylation of STAT3 5.6- to 6.0-fold and of STAT1 4.0-fold in adipose tissue. Leptin tended to increase STAT3 phosphorylation in liver and muscle. Both hormones also increased MAPK phosphorylation: leptin increased it 3.2- to 3.8-fold in adipose tissue and liver, whereas insulin stimulated MAPK phosphorylation 5.0-fold in adipose tissue, 6.8-fold in liver, and 2.5-fold in muscle. Leptin was much less effective than insulin at stimulating IRS pathways. Leptin increased IRS-1-associated PI 3-kinase activity in adipose tissue only 2.0-fold (P < 0.01) compared with the 10-fold effect of insulin. IRS-2-associated PI 3-kinase activity was increased 1.7-fold (P < 0.01) by leptin in liver and 6-fold by insulin. Akt phosphorylation and activity were not changed by leptin but increased with insulin. Lower concentrations of leptin (10 and 50 microg/kg) also stimulated STAT3 phosphorylation in fat. These effects appear to be direct because 3 min after leptin intracerebroventricular injection, phosphorylation of STAT3, STAT1, and MAPK were not stimulated in hypothalamus or adipose tissue. Furthermore, leptin activated STAT3 and MAPK in adipose tissue explants ex vivo and in 3T3-L1 adipocytes. Leptin did not activate STAT3 or MAPK in adipose tissue of db/db mice. Thus, leptin rapidly activates signaling pathways directly at the level of insulin sensitive tissues through the long-form leptin receptor, and these pathways overlap with, but are distinct from, those engaged by insulin.


Subject(s)
Insulin/physiology , Leptin/pharmacology , Protein Serine-Threonine Kinases , Signal Transduction/drug effects , 3T3 Cells , Adipose Tissue/metabolism , Animals , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Injections, Intraventricular , Liver/metabolism , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Rats , Rats, Sprague-Dawley , STAT1 Transcription Factor , STAT3 Transcription Factor , Trans-Activators/metabolism
19.
J Clin Invest ; 105(12): 1827-32, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10862798

ABSTRACT

Obesity in humans and in rodents is usually associated with high circulating leptin levels and leptin resistance. To examine the molecular basis for leptin resistance, we determined the ability of leptin to induce hypothalamic STAT3 (signal transducer and activator of transcription) signaling in C57BL/6J mice fed either low-fat or high-fat diets. In mice fed the low-fat diet, leptin activated STAT3 signaling when administered via the intraperitoneal (ip) or the intracerebroventricular (icv) route, with the half-maximal dose being 30-fold less when given by the icv route. The high-fat diet increased body-weight gain and plasma leptin levels. After 4 weeks on the diet, hypothalamic STAT3 signaling after ip leptin administration was equivalent in both diet groups. In contrast, peripherally administered leptin was completely unable to activate hypothalamic STAT3 signaling, as measured by gel shift assay after 15 weeks of high-fat diet. Despite the absence of detectable signaling after peripheral leptin at 15 weeks, the mice fed the high-fat diet retained the capacity to respond to icv leptin, although the magnitude of STAT3 activation was substantially reduced. These results suggest that leptin resistance induced by a high-fat diet evolves during the course of the diet and has at least two independent causes: an apparent defect in access to sites of action in the hypothalamus that markedly limits the ability of peripheral leptin to activate hypothalamic STAT signaling, and an intracellular signaling defect in leptin-responsive hypothalamic neurons that lies upstream of STAT3 activation.


Subject(s)
Dietary Fats , Hypothalamus/physiology , Leptin/pharmacology , Leptin/physiology , Obesity/physiopathology , Animals , Cerebral Ventricles/drug effects , Cerebral Ventricles/physiology , Cerebral Ventricles/physiopathology , DNA-Binding Proteins/metabolism , Diet, Fat-Restricted , Drug Resistance , Humans , Hypothalamus/drug effects , Hypothalamus/physiopathology , Injections, Intraventricular , Leptin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Obesity/blood , STAT3 Transcription Factor , Trans-Activators/metabolism
20.
Annu Rev Physiol ; 62: 413-37, 2000.
Article in English | MEDLINE | ID: mdl-10845097

ABSTRACT

The discovery of the adipose-derived hormone leptin has generated enormous interest in the interaction between peripheral signals and brain targets involved in the regulation of feeding and energy balance. Plasma leptin levels correlate with fat stores and respond to changes in energy balance. It was initially proposed that leptin serves a primary role as an anti-obesity hormone, but this role is commonly thwarted by leptin resistance. Leptin also serves as a mediator of the adaptation to fasting, and this role may be the primary function for which the molecule evolved. There is increasing evidence that leptin has systemic effects apart from those related to energy homeostasis, including regulation of neuroendocrine and immune function and a role in development.


Subject(s)
Leptin/physiology , Adipose Tissue/physiology , Animals , Humans , Leptin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL