Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
Genes Dev ; 29(9): 934-47, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25934505

ABSTRACT

MAF1 is a global repressor of RNA polymerase III transcription that regulates the expression of highly abundant noncoding RNAs in response to nutrient availability and cellular stress. Thus, MAF1 function is thought to be important for metabolic economy. Here we show that a whole-body knockout of Maf1 in mice confers resistance to diet-induced obesity and nonalcoholic fatty liver disease by reducing food intake and increasing metabolic inefficiency. Energy expenditure in Maf1(-/-) mice is increased by several mechanisms. Precursor tRNA synthesis was increased in multiple tissues without significant effects on mature tRNA levels, implying increased turnover in a futile tRNA cycle. Elevated futile cycling of hepatic lipids was also observed. Metabolite profiling of the liver and skeletal muscle revealed elevated levels of many amino acids and spermidine, which links the induction of autophagy in Maf1(-/-) mice with their extended life span. The increase in spermidine was accompanied by reduced levels of nicotinamide N-methyltransferase, which promotes polyamine synthesis, enables nicotinamide salvage to regenerate NAD(+), and is associated with obesity resistance. Consistent with this, NAD(+) levels were increased in muscle. The importance of MAF1 for metabolic economy reveals the potential for MAF1 modulators to protect against obesity and its harmful consequences.


Subject(s)
Repressor Proteins/genetics , Animals , Autophagy/genetics , Eating/genetics , Energy Metabolism/genetics , Lipid Metabolism/genetics , Longevity/genetics , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Obesity/genetics , RNA, Transfer/metabolism , Spermidine/metabolism
2.
PLoS Biol ; 16(4): e2004399, 2018 04.
Article in English | MEDLINE | ID: mdl-29689050

ABSTRACT

Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) respond to numerous hormonal and neural signals, resulting in changes in food intake. Here, we demonstrate that ARC POMC neurons express capsaicin-sensitive transient receptor potential vanilloid 1 receptor (TRPV1)-like receptors. To show expression of TRPV1-like receptors in ARC POMC neurons, we use single-cell reverse transcription-polymerase chain reaction (RT-PCR), immunohistochemistry, electrophysiology, TRPV1 knock-out (KO), and TRPV1-Cre knock-in mice. A small elevation of temperature in the physiological range is enough to depolarize ARC POMC neurons. This depolarization is blocked by the TRPV1 receptor antagonist and by Trpv1 gene knockdown. Capsaicin-induced activation reduces food intake that is abolished by a melanocortin receptor antagonist. To selectively stimulate TRPV1-like receptor-expressing ARC POMC neurons in the ARC, we generate an adeno-associated virus serotype 5 (AAV5) carrying a Cre-dependent channelrhodopsin-2 (ChR2)-enhanced yellow fluorescent protein (eYFP) expression cassette under the control of the two neuronal POMC enhancers (nPEs). Optogenetic stimulation of TRPV1-like receptor-expressing POMC neurons decreases food intake. Hypothalamic temperature is rapidly elevated and reaches to approximately 39 °C during treadmill running. This elevation is associated with a reduction in food intake. Knockdown of the Trpv1 gene exclusively in ARC POMC neurons blocks the feeding inhibition produced by increased hypothalamic temperature. Taken together, our findings identify a melanocortinergic circuit that links acute elevations in hypothalamic temperature with acute reductions in food intake.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Eating/genetics , Neurons/metabolism , Pro-Opiomelanocortin/genetics , TRPV Cation Channels/genetics , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/drug effects , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Capsaicin/pharmacology , Channelrhodopsins/genetics , Channelrhodopsins/metabolism , Eating/drug effects , Enhancer Elements, Genetic , Female , Gene Expression Regulation , Gene Knock-In Techniques , Gene Knockout Techniques , Genes, Reporter , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Transgenic , Neurons/cytology , Neurons/drug effects , Optogenetics , Physical Conditioning, Animal , Pro-Opiomelanocortin/metabolism , Receptors, Melanocortin/genetics , Receptors, Melanocortin/metabolism , Signal Transduction , Single-Cell Analysis , TRPV Cation Channels/agonists , TRPV Cation Channels/deficiency , Temperature
3.
J Immunol ; 201(7): 1837-1841, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30135180

ABSTRACT

Various malignancies are reproducibly cured in mouse models, but most cancer immunotherapies show objective responses in a fraction of treated patients. One reason for this disconnect may be the use of young, lean mice lacking immune-altering comorbidities present in cancer patients. Although many cancer patients are overweight or obese, the effect of obesity on antitumor immunity is understudied in preclinical tumor models. We examined the effect of obesity on two immunotherapeutic models: systemic anti-CTLA-4 mAb and intratumoral delivery of a TRAIL-encoding adenovirus plus CpG. Both therapies were effective in lean mice, but neither provided a survival benefit to diet-induced obese BALB/c mice. Interestingly, tumor-bearing leptin-deficient (ob/ob) obese BALB/c mice did respond to treatment. Moreover, reducing systemic leptin with soluble leptin receptor:Fc restored the antitumor response in diet-induced obese mice. These data demonstrate the potential of targeting leptin to improve tumor immunotherapy when immune-modulating comorbidities are present.


Subject(s)
Adenocarcinoma/metabolism , Aging/physiology , Antibodies, Monoclonal/therapeutic use , Immunotherapy/methods , Kidney Neoplasms/metabolism , Leptin/metabolism , Obesity/metabolism , Adenocarcinoma/therapy , Adenoviridae/genetics , Animals , CTLA-4 Antigen/immunology , Cell Line, Tumor , Diet , Disease Models, Animal , Female , Humans , Immunity , Kidney Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Obesity/therapy , Oligodeoxyribonucleotides/metabolism , Receptors, Fc/genetics , Receptors, Fc/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , TNF-Related Apoptosis-Inducing Ligand/genetics , TNF-Related Apoptosis-Inducing Ligand/metabolism
4.
bioRxiv ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38659949

ABSTRACT

Background and Aims: The visceral organ-brain axis, mediated by vagal sensory neurons in the vagal nerve ganglion, is essential for maintaining various physiological functions. In this study, we investigated the impact of liver-projecting vagal sensory neurons on energy balance, hepatic steatosis, and anxiety-like behavior in mice under obesogenic conditions. Methods: We performed single-nucleus RNA sequencing of vagal sensory neurons innervating the liver. Based on our snRNA-Seq results, we used the Avil CreERT2 strain to identify vagal sensory neurons that innervate the liver. Results: A small subset of polymodal sensory neurons innervating the liver was located in the left and right ganglia, projecting centrally to the nucleus of the tractus solitarius, area postrema, and dorsal motor nucleus of the vagus, and peripherally to the periportal areas in the liver. Male and female control mice developed diet-induced obesity (DIO) during high-fat diet feeding. Deleting liver-projecting advillin-positive vagal sensory neurons prevented DIO in male and female mice, and these outcomes are associated with increased energy expenditure. Although males and females exhibited improved glucose homeostasis following disruption of liver-projecting vagal sensory neurons, only male mice displayed increased insulin sensitivity. The loss of liver-projecting vagal sensory neurons limited the progression of hepatic steatosis in male and female mice fed a steatogenic diet. Finally, mice lacking liver-innervating vagal sensory neurons exhibited less anxiety-like behavior compared to the control mice. Conclusions: The liver-brain axis contributes to the regulation of energy balance, glucose tolerance, hepatic steatosis, and anxiety-like behavior depending on the nutrient status in healthy and obesogenic conditions.

5.
Am J Physiol Endocrinol Metab ; 305(12): E1512-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24169048

ABSTRACT

Deficient leptin signaling causes infertility via reduced activity of GnRH neurons, causing a hypogonadal state in both rodents and humans. Because GnRH neurons do not express leptin receptors, leptin's effect on GnRH neurons must be indirect. Neurons within the hypothalamic arcuate nucleus that coexpress AGRP and NPY are considered to be important intermediate neurons involved in leptin regulation of GnRH neurons. Previously, we reported that the absence of AGRP and haploinsufficiency of MC4R in leptin receptor mutant (Lepr(db/db)) females result in restoration of fertility and lactation despite the persistence of obesity and insulin resistance. The overarching hypothesis in the present study is that the absence or reduction of leptin's inhibition of AGRP/NPY neurons leads to suppression of GnRH release in cases of leptin signaling deficiency. Since TAC2 (NKB)-TAC3R signaling plays a role in puberty maturation and is modulated by metabolic status, the other aim of this study is to test whether TAC2/NKB neurons in ARC regulated by melanocortinergic signals herein affect leptin's action on puberty and reproduction. Our data showed that AGRP deficiency in Lepr(db/db) females restores normal timing of vaginal opening and estrous cycling, although uterine weight gain and mammary gland development are morphologically delayed. Nonetheless, Agrp(-/-) Lepr(db/db) females are fertile and sustain adequate nutrition of pups with lactation to weaning age. AGRP deficiency results in advanced vaginal opening in wild-type female mice. The postpubertal increase in hypothalamic TAC2 mRNA was not observed in Lepr(db/db) females, whereas AGRP deficiency restored it in Lepr(db/db) females. Additionally, MC4R activation with MTII induced FOS expression in TAC2 neurons, supporting the concept of melanocortinergic regulation of TAC2 neurons. These studies suggest that AGRP imposes an inhibitory effect on puberty and that TAC2 neurons may transmit melanocortinergic inhibition of GnRH neurons.


Subject(s)
Agouti-Related Protein/physiology , Leptin/pharmacology , Receptors, Leptin/physiology , Reproduction , Sexual Maturation , Animals , Body Composition/drug effects , Body Composition/genetics , Estrous Cycle/drug effects , Estrous Cycle/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Reproduction/drug effects , Reproduction/genetics , Sex Factors , Sexual Maturation/drug effects , Sexual Maturation/genetics
6.
Int J Neuropsychopharmacol ; 16(4): 857-67, 2013 May.
Article in English | MEDLINE | ID: mdl-22932068

ABSTRACT

Previous studies have demonstrated that leptin and its receptors (LepRb) in the central nervous system play an important role in regulating depression- and anxiety-related behaviours. However, the physiological functions of LepRb in specific brain regions for mediating different emotional behaviours remain to be defined. In this study, we examined the behavioural effects of LepRb ablation in the adult hippocampus using a series of behavioural paradigms for assessing depression- and anxiety-related behaviours. Targeted deletion of LepRb was achieved using the Cre/loxP site-specific recombination system through bilateral stereotaxic delivery of an adeno-associated virus expressing Cre-recombinase (AAV-Cre) into the dentate gyrus of adult mice homozygous for a floxed leptin receptor allele. AAV-Cre-mediated deletion of the floxed region of LepRb was detected 2 wk after injection. In accordance with this, leptin-stimulated phosphorylation of Akt was attenuated in the hippocampus of AAV-Cre injected mice. Mice injected with AAV-Cre displayed normal locomotor activity and anxiety-like behaviour, as determined in the elevated plus-maze, light-dark box and open field tests, but showed increased depression-like behaviours in the tail suspension, saccharin preference and learned helplessness tests. Taken together, these data suggest that deletion of LepRb in the adult hippocampus is sufficient to induce depression-like behaviours. Our results support the view that leptin signalling in the hippocampus may be essential for positive mood states and active coping to stress.


Subject(s)
Depression/metabolism , Eating/physiology , Eating/psychology , Hippocampus/metabolism , Motor Activity/physiology , Receptors, Leptin/deficiency , Age Factors , Animals , Depression/genetics , Depression/psychology , Eating/genetics , Hippocampus/physiopathology , Male , Mice , Mice, Knockout , Motor Activity/genetics , Receptors, Leptin/genetics
7.
Nat Med ; 12(5): 534-40, 2006 May.
Article in English | MEDLINE | ID: mdl-16604086

ABSTRACT

Leptin controls food intake by regulating the transcription of key neuropeptides in the hypothalamus. The mechanism by which leptin regulates gene expression is unclear, however. Here we show that delivery of adenovirus encoding a constitutively nuclear mutant FoxO1, a transcription factor known to control liver metabolism and pancreatic beta-cell function, to the hypothalamic arcuate nucleus of rodents results in a loss of the ability of leptin to curtail food intake and suppress expression of Agrp. Conversely, a transactivation-deficient FoxO1 mutant prevents induction of Agrp by fasting. We also find that FoxO1 and the transcription factor Stat3 exert opposing actions on the expression of Agrp and Pomc through transcriptional squelching. FoxO1 promotes opposite patterns of coactivator-corepressor exchange at the Pomc and Agrp promoters, resulting in activation of Agrp and inhibition of Pomc. Thus, FoxO1 represents a shared component of pathways integrating food intake and peripheral metabolism.


Subject(s)
Eating , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Leptin/metabolism , Proteins/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Agouti-Related Protein , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Cells, Cultured , Fasting , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Transfer Techniques , Intercellular Signaling Peptides and Proteins , Mice , Neurons/cytology , Neurons/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Promoter Regions, Genetic , Proteins/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
8.
J Infect Dis ; 205(5): 830-40, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22293433

ABSTRACT

Brown adipose tissue (BAT) and white adipose tissue (WAT) and adipocytes are targets of Trypanosoma cruzi infection. Adipose tissue obtained from CD-1 mice 15 days after infection, an early stage of infection revealed a high parasite load. There was a significant increase in macrophages in infected adipose tissue and a reduction in lipid accumulation, adipocyte size, and fat mass and increased expression of lipolytic enzymes. Infection increased levels of Toll-like receptor (TLR) 4 and TLR9 and in the expression of components of the mitogen-activated protein kinase pathway. Protein and messenger RNA (mRNA) levels of peroxisome proliferator-activated receptor γ were increased in WAT, whereas protein and mRNA levels of adiponectin were significantly reduced in BAT and WAT. The mRNA levels of cytokines, chemokines, and their receptors were increased. Nuclear Factor Kappa B levels were increased in BAT, whereas Iκκ-γ levels increased in WAT. Adipose tissue is an early target of T. cruzi infection.


Subject(s)
Adipocytes/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Chagas Disease/parasitology , RNA, Messenger/metabolism , Signal Transduction , Trypanosoma cruzi , Adipocytes/parasitology , Adipocytes/pathology , Adiponectin/metabolism , Adipose Tissue, Brown/parasitology , Adipose Tissue, Brown/pathology , Adipose Tissue, White/parasitology , Adipose Tissue, White/pathology , Animals , Chagas Disease/pathology , Chemokines/metabolism , Cytokines , I-kappa B Kinase/metabolism , Male , Mice , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , PPAR gamma/metabolism , Receptors, Chemokine/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism
9.
Cell Metab ; 4(4): 291-302, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17011502

ABSTRACT

The hormone leptin plays a crucial role in maintenance of body weight and glucose homeostasis. This occurs through central and peripheral pathways, including regulation of insulin secretion by pancreatic beta cells. To study this further in mice, we disrupted the signaling domain of the leptin receptor gene in beta cells and hypothalamus. These mice develop obesity, fasting hyperinsulinemia, impaired glucose-stimulated insulin release, and glucose intolerance, similar to leptin receptor null mice. However, whereas complete loss of leptin function causes increased food intake, this tissue-specific attenuation of leptin signaling does not alter food intake or satiety responses to leptin. Moreover, unlike other obese models, these mice have reduced fasting blood glucose. These results indicate that leptin regulation of glucose homeostasis extends beyond insulin sensitivity to influence beta cell function, independent of pathways controlling food intake. These data suggest that defects in this adipoinsular axis could contribute to diabetes associated with obesity.


Subject(s)
Glucose/metabolism , Homeostasis , Insulin-Secreting Cells/metabolism , Leptin/metabolism , Animals , Eating , Female , Glucose/pharmacology , Insulin/metabolism , Insulin Secretion , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Receptors, Leptin , Sensitivity and Specificity , Signal Transduction/physiology , Time Factors
10.
Am J Pathol ; 177(6): 3133-44, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21056997

ABSTRACT

Obesity represents a risk factor for certain types of cancer. Leptin, a hormone predominantly produced by adipocytes, is elevated in the obese state. In the context of breast cancer, leptin derived from local adipocytes is present at high concentrations within the mammary gland. A direct physiological role of peripheral leptin action in the tumor microenvironment in vivo has not yet been examined. Here, we report that mice deficient in the peripheral leptin receptor, while harboring an intact central leptin signaling pathway, develop a fully mature ductal epithelium, a phenomenon not observed in db/db mice to date. In the context of the MMTV-PyMT mammary tumor model, the lack of peripheral leptin receptors attenuated tumor progression and metastasis through a reduction of the ERK1/2 and Jak2/STAT3 pathways. These are tumor cell-autonomous properties, independent of the metabolic state of the host. In the absence of leptin receptor signaling, the metabolic phenotype is less reliant on aerobic glycolysis and displays an enhanced capacity for ß-oxidation, in contrast to nontransformed cells. Leptin receptor-free tumor cells display reduced STAT3 tyrosine phosphorylation on residue Y705 but have increased serine phosphorylation on residue S727, consistent with preserved mitochondrial function in the absence of the leptin receptor. Therefore, local leptin action within the mammary gland is a critical mediator, linking obesity and dysfunctional adipose tissue with aggressive tumor growth.


Subject(s)
Mitochondria/physiology , Neoplasms/metabolism , Receptors, Leptin/physiology , Animals , Cell Proliferation/drug effects , Cell Respiration/genetics , Down-Regulation/genetics , Female , Leptin/metabolism , Leptin/pharmacology , Leptin/physiology , Male , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Mutant Strains , Mice, Transgenic , Mitochondria/metabolism , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/pathology , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction/genetics , Signal Transduction/physiology , Tumor Cells, Cultured
11.
J Infect Dis ; 202(7): 1104-13, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20726767

ABSTRACT

Mice carrying a defective leptin receptor gene (db/db mice) are metabolically challenged and upon infection with Trypanosoma cruzi (Brazil strain) suffer high mortality. In genetically modified db/db mice, (NSE-Rb db/db mice), central leptin signaling is reconstituted only in the brain, which is sufficient to correct the metabolic defects. NSE-Rb db/db mice were infected with T. cruzi to determine the impact of the lack of leptin signaling on infection in the absence of metabolic dysregulation. Parasitemia levels, mortality rates, and tissue parasitism were statistically significantly increased in infected db/db mice compared with those in infected NSE-Rb db/db and FVB wild-type mice. There was a reduction in fat mass and blood glucose level in infected db/db mice. Plasma levels of several cytokines and chemokines were statistically significantly increased in infected db/db mice compared with those in infected FVB and NSE-Rb db/db mice. These findings suggest that leptin resistance in individuals with obesity and diabetes mellitus may have adverse consequences in T. cruzi infection.


Subject(s)
Chagas Disease/pathology , Chagas Disease/parasitology , Receptors, Leptin/physiology , Trypanosoma cruzi/pathogenicity , Adipose Tissue/pathology , Animals , Blood Glucose/analysis , Chagas Disease/mortality , Cytokines/blood , Mice , Parasitemia , Receptors, Leptin/deficiency , Survival Analysis
12.
Dev Cell ; 56(6): 747-760.e6, 2021 03 22.
Article in English | MEDLINE | ID: mdl-33667344

ABSTRACT

Loss of insulin-secreting pancreatic ß cells through apoptosis contributes to the progression of type 2 diabetes, but underlying mechanisms remain elusive. Here, we identify a pathway in which the cell death inhibitor ARC paradoxically becomes a killer during diabetes. While cytoplasmic ARC maintains ß cell viability and pancreatic architecture, a pool of ARC relocates to the nucleus to induce ß cell apoptosis in humans with diabetes and several pathophysiologically distinct mouse models. ß cell death results through the coordinate downregulation of serpins (serine protease inhibitors) not previously known to be synthesized and secreted by ß cells. Loss of the serpin α1-antitrypsin from the extracellular space unleashes elastase, triggering the disruption of ß cell anchorage and subsequent cell death. Administration of α1-antitrypsin to mice with diabetes prevents ß cell death and metabolic abnormalities. These data uncover a pathway for ß cell loss in type 2 diabetes and identify an FDA-approved drug that may impede progression of this syndrome.


Subject(s)
Apoptosis , Cell Nucleus/metabolism , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/pathology , Insulin-Secreting Cells/pathology , Nerve Tissue Proteins/metabolism , alpha 1-Antitrypsin/chemistry , Animals , Apoptosis Regulatory Proteins/physiology , Cytoplasm/metabolism , Cytoskeletal Proteins/genetics , Diabetes Mellitus, Experimental/etiology , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Type 2/etiology , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Proteins/physiology , Nerve Tissue Proteins/genetics , alpha 1-Antitrypsin/genetics , alpha 1-Antitrypsin/metabolism
13.
Cell Metab ; 1(1): 63-72, 2005 Jan.
Article in English | MEDLINE | ID: mdl-16054045

ABSTRACT

Leptin is required for normal energy and glucose homeostasis. The hypothalamic arcuate nucleus (ARH) has been proposed as an important site of leptin action. To assess the physiological significance of leptin signaling in the ARH, we used mice homozygous for a FLPe-reactivatable, leptin receptor null allele (Lepr(neo/neo) mice). Similar to Lepr(db/db) mice, these mice are obese, hyperglycemic, hyperinsulinemic, infertile, and hypoactive. To selectively restore leptin signaling in the ARH, we generated an adeno-associated virus expressing FLPe-recombinase, which was delivered unilaterally into the hypothalamus using stereotaxic injections. We found that unilateral restoration of leptin signaling in the ARH of Lepr(neo/neo) mice leads to a modest decrease in body weight and food intake. In contrast, unilateral reactivation markedly improved hyperinsulinemia and normalized blood glucose levels and locomotor activity. These data demonstrate that leptin signaling in the ARH is sufficient for mediating leptin's effects on glucose homeostasis and locomotor activity.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Glucose/metabolism , Hypothalamus/metabolism , Leptin/metabolism , Alleles , Animals , Body Composition , Body Weight , Cell Nucleus/metabolism , DNA Nucleotidyltransferases/metabolism , Fertility , Green Fluorescent Proteins/metabolism , Homeostasis , Homozygote , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Genetic , Motor Activity , Neurons/metabolism , Oxygen Consumption , Receptors, Cell Surface/metabolism , Receptors, Leptin , Signal Transduction , Time Factors
14.
J Lipid Res ; 51(7): 1929-42, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20164094

ABSTRACT

Fat is delivered to tissues by apoB-containing lipoproteins synthesized in the liver and intestine with the help of an intracellular chaperone, microsomal triglyceride transfer protein (MTP). Leptin, a hormone secreted by adipose tissue, acts in the brain and on peripheral tissues to regulate fat storage and metabolism. Our aim was to identify the role of leptin signaling in MTP regulation and lipid absorption using several mouse models deficient in leptin receptor (LEPR) signaling and downstream effectors. Mice with spontaneous LEPR B mutations or targeted ablation of LEPR B in proopiomelanocortin (POMC) or agouti gene related peptide (AGRP) expressing cells had increased triglyceride in plasma, liver, and intestine. Furthermore, melanocortin 4 receptor (MC4R) knockout mice expressed a similar triglyceride phenotype, suggesting that leptin might regulate intestinal MTP expression through the melanocortin pathway. Mechanistic studies revealed that the accumulation of triglyceride in the intestine might be secondary to decreased expression of MTP and lipid absorption in these mice. Surgical and chemical blockade of vagal efferent outflow to the intestine in wild-type mice failed to alter the triglyceride phenotype, demonstrating that central neural control mechanisms were likely not involved in the observed regulation of intestinal MTP. Instead, we found that enterocytes express LEPR, POMC, AGRP, and MC4R. We propose that a peripheral, local gut signaling mechanism involving LEPR B and MC4R regulates intestinal MTP and controls intestinal lipid absorption.


Subject(s)
Carrier Proteins/metabolism , Intestinal Absorption/physiology , Leptin/metabolism , Lipid Metabolism , Melanocortins/metabolism , Signal Transduction/physiology , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Animals , Carrier Proteins/genetics , Cholesterol/metabolism , Dietary Fats/metabolism , Enterocytes/metabolism , Intestinal Mucosa/metabolism , Intestines/innervation , Leptin/genetics , Liver/innervation , Liver/metabolism , Melanocortins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Neurons/cytology , Neurons/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Triglycerides/metabolism , Vagotomy
15.
Trends Endocrinol Metab ; 31(1): 46-52, 2020 01.
Article in English | MEDLINE | ID: mdl-31629614

ABSTRACT

Obesity is a neurological disorder that operates by favoring energy storage within adipose depots and increased caloric intake. Most cases of human obesity are acquired without any underlying genetic basis. Here, we suggest that obesity can impair the function of some hypothalamic neurons critical to body weight regulation. Genetic ablation of the retinoblastoma (Rb) gene within pro-opiomelanocortin (POMC) neurons leads to death of the neurons and subsequent obesity. The Rb protein (pRb), a key inhibitor of the cell cycle, can also be inactivated by cyclin dependent kinase (CDK)-mediated phosphorylation. Extensive development led to the production of FDA-approved CDK4/6 inhibitors. Based on our own results, we propose that maintaining or re-instating pRb function using CDK4/6 inhibitors are potentially effective treatments of diet-induced obesity (DIO).


Subject(s)
Obesity/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Humans , Hypothalamus/metabolism , Pro-Opiomelanocortin/metabolism , Retinoblastoma/metabolism
16.
Nat Commun ; 11(1): 6295, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33293550

ABSTRACT

The central melanocortin system plays a fundamental role in the control of feeding and body weight. Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARC) also regulate overall glucose homeostasis via insulin-dependent and -independent pathways. Here, we report that a subset of ARC POMC neurons innervate the liver via preganglionic parasympathetic acetylcholine (ACh) neurons in the dorsal motor nucleus of the vagus (DMV). Optogenetic stimulation of this liver-projecting melanocortinergic pathway elevates blood glucose levels that is associated with increased expression of hepatic gluconeogenic enzymes in female and male mice. Pharmacological blockade and knockdown of the melanocortin-4 receptor gene in the DMV abolish this stimulation-induced effect. Activation of melanocortin-4 receptors inhibits DMV cholinergic neurons and optogenetic inhibition of liver-projecting parasympathetic cholinergic fibers increases blood glucose levels. This elevated blood glucose is not due to altered pancreatic hormone release. Interestingly, insulin-induced hypoglycemia increases ARC POMC neuron activity. Hence, this liver-projecting melanocortinergic circuit that we identified may play a critical role in the counterregulatory response to hypoglycemia.


Subject(s)
Blood Glucose/metabolism , Hypoglycemia/etiology , Liver/innervation , Pro-Opiomelanocortin/metabolism , Vagus Nerve/metabolism , Acetylcholine/metabolism , Action Potentials/physiology , Animals , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/metabolism , Blood Glucose/analysis , Cholinergic Neurons/metabolism , Corticosterone/blood , Corticosterone/metabolism , Disease Models, Animal , Efferent Pathways/physiology , Female , Gene Knockdown Techniques , Glucagon/blood , Glucagon/metabolism , Gluconeogenesis/genetics , Humans , Hypoglycemia/blood , Hypoglycemia/diagnosis , Insulin/blood , Insulin/metabolism , Liver/enzymology , Male , Mice , Optogenetics , RNA, Messenger/metabolism , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Up-Regulation , Vagus Nerve/cytology
17.
Neuron ; 48(6): 1055-66, 2005 Dec 22.
Article in English | MEDLINE | ID: mdl-16364907

ABSTRACT

Recently developed therapeutics for obesity, targeted against cannabinoid receptors, result in decreased appetite and sustained weight loss. Prior studies have demonstrated CB1 receptors (CB1Rs) and leptin modulation of cannabinoid synthesis in hypothalamic neurons. Here, we show that depolarization of perifornical lateral hypothalamus (LH) neurons elicits a CB1R-mediated suppression of inhibition in local circuits thought to be involved in appetite and "natural reward." The depolarization-induced decrease in inhibitory tone to LH neurons is blocked by leptin. Leptin inhibits voltage-gated calcium channels in LH neurons via the activation of janus kinase 2 (JAK2) and of mitogen-activated protein kinase (MAPK). Leptin-deficient mice are characterized by both an increase in steady-state voltage-gated calcium currents in LH neurons and a CB1R-mediated depolarization-induced suppression of inhibition that is 6-fold longer than that in littermate controls. Our data provide direct electrophysiological support for the involvement of endocannabinoids and leptin as modulators of hypothalamic circuits underlying motivational aspects of feeding behavior.


Subject(s)
Appetite Regulation/physiology , Cannabinoid Receptor Modulators/metabolism , Endocannabinoids , Hypothalamic Area, Lateral/metabolism , Leptin/metabolism , Neural Pathways/metabolism , Signal Transduction/physiology , Animals , Appetite Regulation/drug effects , Calcium Channels/drug effects , Calcium Channels/metabolism , Calcium Signaling/drug effects , Calcium Signaling/genetics , Cannabinoid Receptor Modulators/pharmacology , Hypothalamic Area, Lateral/drug effects , Hypothalamus/drug effects , Hypothalamus/physiology , Janus Kinase 2 , Leptin/genetics , Leptin/pharmacology , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motivation , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neural Pathways/drug effects , Neurons/drug effects , Neurons/metabolism , Organ Culture Techniques , Patch-Clamp Techniques , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptors, Leptin , Reward , Signal Transduction/drug effects , Time Factors
18.
Mol Endocrinol ; 22(8): 1950-61, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18511494

ABSTRACT

The nuclear receptor steroidogenic factor 1 (SF-1) plays essential roles in the development and function of the ventromedial hypothalamic nucleus (VMH). Considerable evidence links the VMH and SF-1 with the regulation of energy homeostasis. Here, we demonstrate that SF-1 colocalizes in VMH neurons with the cannabinoid receptor 1 (CB1R) and that a specific CB1R agonist modulates electrical activity of SF-1 neurons in hypothalamic slice preparations. We further show that SF-1 directly regulates CB1R gene expression via a SF-1-responsive element at -101 in its 5'-flanking region. Finally, we show that knockout mice with selective inactivation of SF-1 in the brain have decreased expression of CB1R in the region of the VMH and exhibit a blunted response to systemically administered CB1R agonists. These studies suggest that SF-1 directly regulates the expression of CB1R, which has been implicated in the regulation of energy homeostasis and anxiety-like behavior.


Subject(s)
Gene Expression Regulation , Receptor, Cannabinoid, CB1/genetics , Steroidogenic Factor 1/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Action Potentials/drug effects , Animals , Arachidonic Acids/administration & dosage , Arachidonic Acids/pharmacology , Body Weight/drug effects , Cell Line , Cells, Cultured , Darkness , Electrophysiology , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Feeding Behavior/drug effects , Gene Expression Regulation/drug effects , Humans , In Vitro Techniques , Mice , Mice, Knockout , Neurons/drug effects , Neurons/physiology , Phosphorylation/drug effects , Receptor, Cannabinoid, CB1/metabolism , Response Elements , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/enzymology
19.
Neuron ; 42(6): 983-91, 2004 Jun 24.
Article in English | MEDLINE | ID: mdl-15207242

ABSTRACT

Neuroanatomical and electrophysiological studies have shown that hypothalamic POMC neurons are targets of the adipostatic hormone leptin. However, the physiological relevance of leptin signaling in these neurons has not yet been directly tested. Here, using the Cre/loxP system, we critically test the functional importance of leptin action on POMC neurons by deleting leptin receptors specifically from these cells in mice. Mice lacking leptin signaling in POMC neurons are mildly obese, hyperleptinemic, and have altered expression of hypothalamic neuropeptides. In summary, leptin receptors on POMC neurons are required but not solely responsible for leptin's regulation of body weight homeostasis.


Subject(s)
Body Weight/physiology , Homeostasis/physiology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Receptors, Cell Surface/physiology , Signal Transduction/physiology , Age Factors , Animals , Body Composition/genetics , Eating/genetics , Female , Gene Expression/genetics , Green Fluorescent Proteins , Hypothalamus/cytology , Hypothalamus/metabolism , Immunohistochemistry/methods , In Situ Hybridization/methods , Leptin/blood , Leptin/pharmacology , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neuropeptides/metabolism , Oxygen Consumption/genetics , Pro-Opiomelanocortin/genetics , RNA, Messenger/metabolism , Receptors, Leptin , Repressor Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Sex Factors , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins , Transcription Factors/metabolism , alpha-MSH/metabolism
20.
Endocrinology ; 149(4): 1773-85, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18162515

ABSTRACT

Two known types of leptin-responsive neurons reside within the arcuate nucleus: the agouti gene-related peptide (AgRP)/neuropeptide Y (NPY) neuron and the proopiomelanocortin (POMC) neuron. By deleting the leptin receptor gene (Lepr) specifically in AgRP/NPY and/or POMC neurons of mice, we examined the several and combined contributions of these neurons to leptin action. Body weight and adiposity were increased by Lepr deletion from AgRP and POMC neurons individually, and simultaneous deletion in both neurons (A+P LEPR-KO mice) further increased these measures. Young (periweaning) A+P LEPR-KO mice exhibit hyperphagia and decreased energy expenditure, with increased weight gain, oxidative sparing of triglycerides, and increased fat accumulation. Interestingly, however, many of these abnormalities were attenuated in adult animals, and high doses of leptin partially suppress food intake in the A+P LEPR-KO mice. Although mildly hyperinsulinemic, the A+P LEPR-KO mice displayed normal glucose tolerance and fertility. Thus, AgRP/NPY and POMC neurons each play mandatory roles in aspects of leptin-regulated energy homeostasis, high leptin levels in adult mice mitigate the importance of leptin-responsiveness in these neurons for components of energy balance, suggesting the presence of other leptin-regulated pathways that partially compensate for the lack of leptin action on the POMC and AgRP/NPY neurons.


Subject(s)
Agouti-Related Protein/physiology , Eating , Energy Metabolism , Pro-Opiomelanocortin/physiology , Receptors, Leptin/physiology , Animals , Body Composition , Fertility , Hyperinsulinism/etiology , Hyperphagia , Lactation , Male , Mice , Neuropeptide Y/physiology
SELECTION OF CITATIONS
SEARCH DETAIL