Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
PeerJ ; 12: e16739, 2024.
Article in English | MEDLINE | ID: mdl-38282864

ABSTRACT

GPR149 is an orphan receptor about which little is known. Accordingly, in the present study, we mapped the tissue expression of Gpr149 in mice using three complementary approaches: quantitative PCR, in situ hybridization, and a newly generated Gpr149-Cre reporter mouse model. The strongest expressions of Gpr149 were observed in neurons of the islands of Calleja, the ventromedial hypothalamus, and the rostral interpeduncular nucleus. Moderate-to-low expression was also observed in the basal forebrain, striatum, hypothalamus, brainstem, and spinal cord. Some Gpr149 expression was also detected in the primary afferent neurons, enteric neurons, and pituitary endocrine cells. This expression pattern is consistent with the involvement of GPR149 signaling in the regulation of energy balance. To explore the physiological function of GPR149 in vivo, we used CRISPR-Cas9 to generate a global knockout allele with mice lacking Gpr149 exon 1. Preliminary metabolic findings indicated that Gpr149-/- mice partially resist weight gain when fed with a high-fat diet and have greater sensitivity to insulin than control mice. In summary, our data may serve as a resource for future in vivo studies on GPR149 in the context of diet-induced obesity.


Subject(s)
Hypothalamus , Obesity , Receptors, G-Protein-Coupled , Animals , Male , Mice , Diet, High-Fat/adverse effects , Homeostasis/genetics , Hypothalamus/metabolism , Obesity/metabolism , Receptors, G-Protein-Coupled/genetics , Weight Gain
2.
bioRxiv ; 2023 Oct 02.
Article in English | MEDLINE | ID: mdl-37808637

ABSTRACT

There has been a dramatic increase in the identification of non-conical translation and a significant expansion of the protein-coding genome and proteome. Among the strategies used to identify novel small ORFs (smORFs), Ribosome profiling (Ribo-Seq) is the gold standard for the annotation of novel coding sequences by reporting on smORF translation. In Ribo-Seq, ribosome-protected footprints (RPFs) that map to multiple sites in the genome are computationally removed since they cannot unambiguously be assigned to a specific genomic location, or to a specific transcript in the case of multiple isoforms. Furthermore, RPFs necessarily result in short (25-34 nucleotides) reads, increasing the chance of ambiguous and multi-mapping alignments, such that smORFs that reside in these regions cannot be identified by Ribo-Seq. Here, we show that the inclusion of proteogenomics to create a Ribosome Profiling and Proteogenomics Pipeline (RP3) bypasses this limitation to identify a group of microprotein-encoding smORFs that are missed by current Ribo-Seq pipelines. Moreover, we show that the microproteins identified by RP3 have different sequence compositions from the ones identified by Ribo-Seq-only pipelines, which can affect proteomics identification. In aggregate, the development of RP3 maximizes the detection and confidence of protein-encoding smORFs and microproteins.

3.
Acta Physiol (Oxf) ; 238(1): e13947, 2023 05.
Article in English | MEDLINE | ID: mdl-36755506

ABSTRACT

AIM: Postprandial secretion of the appetite-inhibiting hormones, glucagon-like peptide-1 (GLP-1), and peptide YY are reduced with obesity. It is unclear if the reduced secretion persists following weight loss (WL), if other appetite-inhibiting hormones are also reduced, and if so whether reduced secretion results from intrinsic changes in the gut. METHODS: To address whether WL may restore secretion of GLP-1 and other appetite-inhibiting hormones, we performed a gut perfusion study of the small intestine in diet-induced obese (DIO) rats after WL. A 20% weight loss (means ± SEM (g): 916 ± 53 vs. 703 ± 35, p < 0.01, n = 7) was induced by calorie restriction, and maintained stable for ≥7 days prior to gut perfusion to allow for complete renewal of enteroendocrine cells. Age-matched DIO rats were used as comparator. Several gut hormones were analyzed from the venous effluent, and gene expression was performed on gut tissue along the entire length of the intestine. RESULTS: Secretion of cholecystokinin, gastrin, glucose-dependent insulinotropic peptide, GLP-1, neurotensin, and somatostatin was not affected by WL during basal conditions (p ≥ 0.25) or in response to macronutrients and bile acids (p ≥ 0.14). Glucose absorption was indistinguishable following WL. The expression of genes encoding the studied peptides, macronutrient transporters (glucose, fructose, and di-/tripeptides) and bile acid receptors did also not differ between DIO and WL groups. CONCLUSIONS: These data suggest that the attenuated postprandial responses of GLP-1, as well as reduced responses of other appetite-inhibiting gut hormones, in people living with obesity may persist after weight loss and may contribute to their susceptibility for weight regain.


Subject(s)
Appetite , Caloric Restriction , Rats , Animals , Glucagon-Like Peptide 1/metabolism , Weight Loss , Obesity/metabolism , Intestine, Small , Glucose
4.
Cell Metab ; 35(1): 166-183.e11, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36599300

ABSTRACT

Microproteins (MPs) are a potentially rich source of uncharacterized metabolic regulators. Here, we use ribosome profiling (Ribo-seq) to curate 3,877 unannotated MP-encoding small ORFs (smORFs) in primary brown, white, and beige mouse adipocytes. Of these, we validated 85 MPs by proteomics, including 33 circulating MPs in mouse plasma. Analyses of MP-encoding mRNAs under different physiological conditions (high-fat diet) revealed that numerous MPs are regulated in adipose tissue in vivo and are co-expressed with established metabolic genes. Furthermore, Ribo-seq provided evidence for the translation of Gm8773, which encodes a secreted MP that is homologous to human and chicken FAM237B. Gm8773 is highly expressed in the arcuate nucleus of the hypothalamus, and intracerebroventricular administration of recombinant mFAM237B showed orexigenic activity in obese mice. Together, these data highlight the value of this adipocyte MP database in identifying MPs with roles in fundamental metabolic and physiological processes such as feeding.


Subject(s)
Adipocytes, White , Adipose Tissue, Brown , Humans , Animals , Mice , Adipocytes, White/metabolism , Adipose Tissue, Brown/metabolism , Open Reading Frames/genetics , Adipose Tissue, White/metabolism , Adipocytes, Brown/metabolism , Micropeptides
5.
Metabolites ; 12(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35050161

ABSTRACT

Obesity is caused by prolonged energy surplus. Current anti-obesity medications are mostly centralized around the energy input part of the energy balance equation by increasing satiety and reducing appetite. Our gastrointestinal tract is a key organ for regulation of food intake and supplies a tremendous number of circulating signals that modulate the activity of appetite-regulating areas of the brain by either direct interaction or through the vagus nerve. Intestinally derived messengers are manifold and include absorbed nutrients, microbial metabolites, gut hormones and other enterokines, collectively comprising a fine-tuned signalling system to the brain. After a meal, nutrients directly interact with appetite-inhibiting areas of the brain and induce satiety. However, overall feeding behaviour also depends on secretion of gut hormones produced by highly specialized and sensitive enteroendocrine cells. Moreover, circulating microbial metabolites and their interactions with enteroendocrine cells further contribute to the regulation of feeding patterns. Current therapies exploiting the appetite-regulating properties of the gut are based on chemically modified versions of the gut hormone, glucagon-like peptide-1 (GLP-1) or on inhibitors of the primary GLP-1 inactivating enzyme, dipeptidyl peptidase-4 (DPP-4). The effectiveness of these approaches shows that that the gut is a promising target for therapeutic interventions to achieve significant weigh loss. We believe that increasing understanding of the functionality of the intestinal epithelium and new delivery systems will help develop selective and safe gut-based therapeutic strategies for improved obesity treatment in the future. Here, we provide an overview of the major homeostatic appetite-regulating signals generated by the intestinal epithelial cells and how these signals may be harnessed to treat obesity by pharmacological means.

6.
Am J Physiol Endocrinol Metab ; 321(4): E443-E452, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34370594

ABSTRACT

Growth differentiating factor 15 (GDF15) is expressed in the intestine and is one of the most recently identified satiety peptides. The mechanisms controlling its secretion are unclear. The present study investigated whether plasma GDF15 concentrations are meal-related and if potential responses depend on macronutrient type or are affected by previous bariatric surgery. The study included 1) volunteers ingesting rapidly vs. slowly digested carbohydrates (sucrose vs. isomaltose; n = 10), 2) volunteers who had undergone Roux-en-Y gastric bypass (RYGB) or sleeve gastrectomy (SG) surgery and unoperated matched controls ingesting a liquid mixed meal (n = 9-10 in each group), and 3) individuals with previous RYGB compared with unoperated controls ingesting isocaloric glucose, fat, or protein (n = 6 in each group). Plasma was collected after an overnight fast and up to 6 h after ingestion (≥12 time points). In cohort 1, fasting GDF15 concentrations were ∼480 pg/mL. Concentrations after sucrose or isomaltose intake did not differ from baseline (P = 0.26 to P > 0.99) and total area under the curves (tAUCs were similar between groups (P = 0.77). In cohort 2, fasting GDF15 concentrations were as follows (pg/mL): RYGB = 540 ± 41.4, SG = 477 ± 36.4, and controls = 590 ± 41.8, with no between-group differences (P = 0.73). Concentrations did not increase at any postprandial time point (over all time factor: P = 0.10) and tAUCs were similar between groups (P = 0.73). In cohort 3, fasting plasma GDF15 was similar among the groups (P > 0.99) and neither glucose, fat, nor protein intake consistently increased the concentrations. In conclusion, we find that plasma GDF15 was not stimulated by meal intake and that fasting concentrations did not differ between RYGB-, SG-, and body mass index (BMI)-matched controls when investigated during the weight stable phase after RYGB and SG.NEW & NOTEWORTHY Our combined data show that GDF15 does not increase in response to a liquid meal. Moreover, we show for the first time that ingestion of sucrose, isomaltose, glucose, fat, or protein also does not increase plasma GDF15 concentrations, questioning the role of GDF15 in regulation of food source preference. Finally, we find that neither fasting nor postprandial plasma GDF15 concentrations are increased in individuals with previous bariatric surgery compared with unoperated body mass index (BMI)-matched controls.


Subject(s)
Bariatric Surgery/methods , Biomarkers/blood , Gastrointestinal Tract/metabolism , Growth Differentiation Factor 15/blood , Meals , Obesity, Morbid/blood , Adult , Blood Glucose/analysis , Body Mass Index , Case-Control Studies , Cross-Over Studies , Female , Follow-Up Studies , Humans , Insulin/blood , Male , Middle Aged , Obesity, Morbid/pathology , Obesity, Morbid/surgery , Postprandial Period , Prognosis , Randomized Controlled Trials as Topic , Weight Loss
7.
Mol Metab ; 53: 101258, 2021 11.
Article in English | MEDLINE | ID: mdl-34023483

ABSTRACT

OBJECTIVE: Acyl-ghrelin regulates eating, body weight, blood glucose, and GH secretion upon binding to its receptor GHSR (growth hormone secretagogue receptor; ghrelin receptor). GHSR is distributed in several brain regions and some peripheral cell-types including pituitary somatotrophs. The objective of the current study was to determine the functional significance of acyl-ghrelin's action on GHSR-expressing somatotrophs in mediating GH secretion and several of acyl-ghrelin's metabolic actions. METHODS: GH-IRES-Cre mice and loxP-flanked (floxed) GHSR mice were newly developed and then crossed to one another to generate mice that lacked GHSR selectively from somatotrophs. Following validation of mice with somatotroph-selective GHSR deletion, metabolic responses of these mice and control littermates were assessed following both acute and chronic acyl-ghrelin administration, a 24-h fast, and a prolonged 60% chronic caloric restriction protocol modeling starvation. RESULTS: In mice with somatotroph-selective GHSR deletion, a single peripheral injection of acyl-ghrelin failed to induce GH secretion or increase food intake, unlike wild-type and other littermate control groups. However, the usual acute blood glucose increase in response to the acyl-ghrelin bolus was preserved. Similarly, chronic s.c. acyl-ghrelin administration to mice with somatotroph-selective GHSR deletion failed to increase plasma GH, food intake, or body weight. Physiologically elevating plasma acyl-ghrelin via a 24-h fast also failed to raise plasma GH and resulted in a limited hyperphagic response upon food reintroduction in mice with somatotroph-selective GHSR deletion, although those mice nonetheless did not exhibit an exaggerated reduction in blood glucose. Physiologically elevating plasma acyl-ghrelin via a 15-day caloric restriction protocol which provided only 40% of usual daily calories failed to raise plasma GH in mice with somatotroph-selective GHSR deletion, although those mice did not exhibit life-threatening hypoglycemia. CONCLUSIONS: These results reveal that direct engagement of GHSR-expressing somatotrophs is required for a peripheral ghrelin bolus to acutely stimulate GH secretion and the actions of chronic acyl-ghrelin delivery and physiological plasma acyl-ghrelin elevations to increase plasma GH. These results also suggest that actions of acyl-ghrelin to increase food intake and body weight are reliant on direct activation of GHSRs expressed on somatotrophs. Furthermore, these results suggest that the glucoregulatory actions of acyl-ghrelin - in particular, its actions to raise blood glucose when acutely administered, prevent small blood glucose drops following a 24-h fast, and avert life-threatening hypoglycemia during an acute-on-chronic caloric restriction protocol - do not depend on GHSR expression by somatotrophs.


Subject(s)
Ghrelin/metabolism , Growth Hormone/metabolism , Animals , Blood Glucose/metabolism , Ghrelin/analogs & derivatives , Mice , Receptors, Ghrelin/deficiency , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism
8.
J Comp Neurol ; 529(1): 111-128, 2021 01.
Article in English | MEDLINE | ID: mdl-32356570

ABSTRACT

While autonomic ganglia have been extensively studied in rats instead of mice, there is renewed interest in the anatomy of the mouse autonomic nervous system. This study examined the prevalence and anatomical features of a cell bridge linking two autonomic ganglia of the neck, namely, the nodose ganglion (NG) and the superior cervical ganglion (SCG) in a cohort of C57BL/6J mice. We identified a cell bridge between the NG and the cranial pole of the SCG. This cell bridge was tubular shaped with an average length and width of 700 and 240 µm, respectively. The cell bridge was frequently unilateral and significantly more prevalent in the ganglionic masses from males (38%) than females (21%). On each of its extremities, it contained a mixed of vagal afferents and postganglionic sympathetic neurons. The two populations of neurons abruptly replaced each other in the middle of the cell bridge. We examined the mRNA expression for selected autonomic markers in samples of the NG with or without cell bridge. Our results indicated that the cell bridge was enriched in both markers of postganglionic sympathetic and vagal afferents neurons. Lastly, using FluoroGold microinjection into the NG, we found that the existence of a cell bridge may occasionally lead to the inadvertent contamination of the SCG. In summary, this study describes the anatomy of a cell bridge variant consisting of the fusion of the mouse NG and SCG. The practical implications of our observations are discussed with respect to studies of the mouse vagal afferents, an area of research of increasing popularity.


Subject(s)
Nodose Ganglion/anatomy & histology , Superior Cervical Ganglion/anatomy & histology , Animals , Female , Male , Mice , Mice, Inbred C57BL , Nodose Ganglion/cytology , Prevalence , Superior Cervical Ganglion/cytology
9.
Sci Adv ; 5(5): eaav4832, 2019 05.
Article in English | MEDLINE | ID: mdl-31149633

ABSTRACT

Ensuring robust gamete production even in the face of environmental stress is of utmost importance for species survival, especially in mammals that have low reproductive rates. Here, we describe a family of genes called melanoma antigens (MAGEs) that evolved in eutherian mammals and are normally restricted to expression in the testis (http://MAGE.stjude.org) but are often aberrantly activated in cancer. Depletion of Mage-a genes disrupts spermatogonial stem cell maintenance and impairs repopulation efficiency in vivo. Exposure of Mage-a knockout mice to genotoxic stress or long-term starvation that mimics famine in nature causes defects in spermatogenesis, decreased testis weights, diminished sperm production, and reduced fertility. Last, human MAGE-As are activated in many cancers where they promote fuel switching and growth of cells. These results suggest that mammalian-specific MAGE genes have evolved to protect the male germline against environmental stress, ensure reproductive success under non-optimal conditions, and are hijacked by cancer cells.


Subject(s)
Melanoma-Specific Antigens/genetics , Neoplasms/genetics , Spermatogenesis/genetics , Stress, Physiological/genetics , Testis/physiology , Animals , DNA Damage , Deoxyglucose/pharmacology , Evolution, Molecular , Female , Gene Expression Regulation, Neoplastic , Germ Cells , Humans , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Spermatogonia/drug effects , Starvation
10.
Cell Rep ; 25(2): 271-277.e4, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30304667

ABSTRACT

Obesity is associated with many complications, including type 2 diabetes and painful neuropathy. There is no cure or prevention for obesity-induced pain, and the neurobiology underlying the onset of the disease is still obscure. In this study, we observe that western diet (WD)-fed mice developed early allodynia with an increase of ER stress markers in the sensory neurons of the dorsal root ganglia (DRG). Using cell-specific approaches, we demonstrate that neuronal liver X receptor (LXR) activation delays ER stress and allodynia in WD-fed mice. Our findings suggest that lipid-binding nuclear receptors expressed in the sensory neurons of the DRG play a role in the onset of obesity-induced hypersensitivity. The LXR and lipid-sensor pathways represent a research avenue to identify targets to prevent debilitating complications affecting the peripheral nerve system in obesity.


Subject(s)
Endoplasmic Reticulum Stress , Ganglia, Spinal/drug effects , Hyperalgesia/etiology , Liver X Receptors/physiology , Obesity/complications , Sensory Receptor Cells/drug effects , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Benzoates/pharmacology , Benzylamines/pharmacology , Diet, Western/adverse effects , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , Hyperalgesia/drug therapy , Hyperalgesia/pathology , Liver X Receptors/agonists , Male , Mice , Mice, Knockout , Sensory Receptor Cells/cytology , Sensory Receptor Cells/metabolism
11.
Mol Metab ; 6(10): 1081-1091, 2017 10.
Article in English | MEDLINE | ID: mdl-29031710

ABSTRACT

OBJECTIVE AND METHODS: Metabolic viscera and their vasculature are richly innervated by peripheral sensory neurons. Here, we examined the metabolic and inflammatory profiles of mice with selective ablation of all Nav1.8-expressing primary afferent neurons. RESULTS: While mice lacking sensory neurons displayed no differences in body weight, food intake, energy expenditure, or body composition compared to controls on chow diet, ablated mice developed an exaggerated inflammatory response to high-fat feeding characterized by bouts of weight loss, splenomegaly, elevated circulating interleukin-6 and hepatic serum amyloid A expression. This phenotype appeared to be directly mediated by the ingestion of saturated lipids. CONCLUSIONS: These data demonstrate that the Nav1.8-expressing afferent neurons are not essential for energy balance but are required for limiting the acute phase response caused by an obesogenic diet.


Subject(s)
Acute-Phase Reaction/metabolism , Dietary Fats/metabolism , NAV1.8 Voltage-Gated Sodium Channel/metabolism , NAV1.8 Voltage-Gated Sodium Channel/physiology , Animals , Body Composition , Body Weight , Diet, High-Fat , Eating/physiology , Energy Metabolism/physiology , Homeostasis/physiology , Mice , Neurons, Afferent/metabolism , Obesity/etiology , Sensory Receptor Cells/metabolism , Viscera/metabolism , Weight Loss
12.
Front Neuroanat ; 9: 120, 2015.
Article in English | MEDLINE | ID: mdl-26388745

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated transcription factor that was originally identified as a regulator of peroxisome proliferation and adipocyte differentiation. Emerging evidence suggests that functional PPARγ signaling also occurs within the hypothalamus. However, the exact distribution and identities of PPARγ-expressing hypothalamic cells remains under debate. The present study systematically mapped PPARγ mRNA expression in the adult mouse brain using in situ hybridization histochemistry. PPARγ mRNA was found to be expressed at high levels outside the hypothalamus including the neocortex, the olfactory bulb, the organ of the vasculosum of the lamina terminalis (VOLT), and the subfornical organ. Within the hypothalamus, PPARγ was present at moderate levels in the suprachiasmatic nucleus (SCh) and the ependymal of the 3rd ventricle. In all examined feeding-related hypothalamic nuclei, PPARγ was expressed at very low levels that were close to the limit of detection. Using qPCR techniques, we demonstrated that PPARγ mRNA expression was upregulated in the SCh in response to fasting. Double in situ hybridization further demonstrated that PPARγ was primarily expressed in neurons rather than glia. Collectively, our observations provide a comprehensive map of PPARγ distribution in the intact adult mouse hypothalamus.

13.
Elife ; 42015 Jun 15.
Article in English | MEDLINE | ID: mdl-26076474

ABSTRACT

Peripheral neural sensory mechanisms play a crucial role in metabolic regulation but less is known about the mechanisms underlying vagal sensing itself. Recently, we identified an enrichment of liver X receptor alpha and beta (LXRα/ß) in the nodose ganglia of the vagus nerve. In this study, we show mice lacking LXRα/ß in peripheral sensory neurons have increased energy expenditure and weight loss when fed a Western diet (WD). Our findings suggest that the ability to metabolize and sense cholesterol and/or fatty acids in peripheral neurons is an important requirement for physiological adaptations to WDs.


Subject(s)
Adaptation, Physiological/physiology , Diet, Western , Energy Metabolism/physiology , Nodose Ganglion/physiology , Orphan Nuclear Receptors/deficiency , Sensory Receptor Cells/metabolism , Analysis of Variance , Animals , Calorimetry, Indirect , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Weight Loss/physiology
14.
Mol Endocrinol ; 29(2): 213-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25495872

ABSTRACT

Hormones such as fibroblast growth factor 21 (FGF21) and glucocorticoids (GCs) play crucial roles in coordinating the adaptive starvation response. Here we examine the interplay between these hormones. It was previously shown that FGF21 induces corticosterone levels in mice by acting on the brain. We now show that this induces the expression of genes required for GC synthesis in the adrenal gland. FGF21 also increases corticosterone secretion from the adrenal in response to ACTH. We further show that the relationship between FGF21 and GCs is bidirectional. GCs induce Fgf21 expression in the liver by acting on the GC receptor (GR). The GR binds in a ligand-dependent manner to a noncanonical GR response element located approximately 4.4 kb upstream of the Fgf21 transcription start site. The GR cooperates with the nuclear fatty acid receptor, peroxisome proliferator-activated receptor-α, to stimulate Fgf21 transcription. GR and peroxisome proliferator-activated receptor-α ligands have additive effects on Fgf21 expression both in vivo and in primary cultures of mouse hepatocytes. We conclude that FGF21 and GCs regulate each other's production in a feed-forward loop and suggest that this provides a mechanism for bypassing negative feedback on the hypothalamic-pituitary-adrenal axis to allow sustained gluconeogenesis during starvation.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucocorticoids/pharmacology , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Base Pairing , Binding Sites , Chromatin/metabolism , Corticosterone/biosynthesis , Dexamethasone/pharmacology , Genetic Loci , Humans , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , PPAR alpha/metabolism , Protein Binding/drug effects , Receptors, Glucocorticoid/metabolism , Transcription Initiation Site , Transcriptional Activation/drug effects
15.
EMBO Mol Med ; 6(12): 1610-21, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25422356

ABSTRACT

The heart requires a continuous supply of energy but has little capacity for energy storage and thus relies on exogenous metabolic sources. We previously showed that cardiac MED13 modulates systemic energy homeostasis in mice. Here, we sought to define the extra-cardiac tissue(s) that respond to cardiac MED13 signaling. We show that cardiac overexpression of MED13 in transgenic (MED13cTg) mice confers a lean phenotype that is associated with increased lipid uptake, beta-oxidation and mitochondrial content in white adipose tissue (WAT) and liver. Cardiac expression of MED13 decreases metabolic gene expression in the heart but enhances them in WAT. Although exhibiting increased energy expenditure in the fed state, MED13cTg mice metabolically adapt to fasting. Furthermore, MED13cTg hearts oxidize fuel that is readily available, rendering them more efficient in the fed state. Parabiosis experiments in which circulations of wild-type and MED13cTg mice are joined, reveal that circulating factor(s) in MED13cTg mice promote enhanced metabolism and leanness. These findings demonstrate that MED13 acts within the heart to promote systemic energy expenditure in extra-cardiac energy depots and point to an unexplored metabolic communication system between the heart and other tissues.


Subject(s)
Adipose Tissue, White/metabolism , Adipose Tissue/metabolism , Liver/metabolism , Mediator Complex/metabolism , Thinness/metabolism , Animals , Energy Metabolism , Female , Humans , Male , Mediator Complex/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/metabolism , Myocardium/metabolism , Signal Transduction , Thinness/genetics
16.
Cell Metab ; 20(4): 670-7, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25130400

ABSTRACT

The mechanism by which pharmacologic administration of the hormone FGF21 increases energy expenditure to cause weight loss in obese animals is unknown. Here we report that FGF21 acts centrally to exert its effects on energy expenditure and body weight in obese mice. Using tissue-specific knockout mice, we show that ßKlotho, the obligate coreceptor for FGF21, is required in the nervous system for these effects. FGF21 stimulates sympathetic nerve activity to brown adipose tissue through a mechanism that depends on the neuropeptide corticotropin-releasing factor. Our findings provide an unexpected mechanistic explanation for the strong pharmacologic effects of FGF21 on energy expenditure and weight loss in obese animals.


Subject(s)
Energy Metabolism/drug effects , Fibroblast Growth Factors/pharmacology , Sympathetic Nervous System/drug effects , Weight Loss/drug effects , Adipose Tissue, Brown/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hypothalamus/metabolism , Klotho Proteins , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , RNA, Messenger/metabolism , Sympathetic Nervous System/metabolism , Thermogenesis/genetics
17.
Cell Metab ; 19(4): 722-30, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24703703

ABSTRACT

The vagus nerve innervates visceral organs providing a link between key metabolic cues and the CNS. However, it is not clear whether vagal neurons can directly respond to changing lipid levels and whether altered "lipid sensing" by the vagus nerve regulates energy balance. In this study, we systematically profiled the expression of all known nuclear receptors in laser-captured nodose ganglion (NG) neurons. In particular, we found PPARγ expression was reduced by high-fat-diet feeding. Deletion of PPARγ in Phox2b neurons promoted HFD-induced thermogenesis that involved the reprograming of white adipocyte into a brown-like adipocyte cell fate. Finally, we showed that PPARγ in NG neurons regulates genes necessary for lipid metabolism and those that are important for synaptic transmission. Collectively, our findings provide insights into how vagal afferents survey peripheral metabolic cues and suggest that the reduction of PPARγ in NG neurons may serve as a protective mechanism against diet-induced weight gain.


Subject(s)
Diet, High-Fat , Lipid Metabolism/physiology , Neurons/metabolism , Nodose Ganglion/cytology , PPAR gamma/metabolism , Thermogenesis/physiology , Adipocytes/cytology , Animals , Cell Differentiation/physiology , Gene Deletion , Gene Expression Regulation/physiology , Laser Capture Microdissection , Lipid Metabolism/genetics , Mice , Mice, Transgenic , Models, Biological , Neurons/physiology , Nodose Ganglion/surgery , PPAR gamma/genetics , Real-Time Polymerase Chain Reaction
18.
EMBO Mol Med ; 6(4): 436-8, 2014 04.
Article in English | MEDLINE | ID: mdl-24623378

ABSTRACT

The heart has been recognized as an endocrine organ for over 30 years (de Bold, 2011); however, little is known about how the heart communicates with other organs in the body, and even less is known about this process in the diseased heart. In this issue of EMBO Molecular Medicine, Magida and Leinwand (2014) introduce the concept that a primary genetic defect in the heart results in aberrant hepatic lipid metabolism, which consequently exacerbates hypertrophic cardiomyopathy (HCM). This study provides evidence in support of the hypothesis that crosstalk occurs between the heart and liver, and that this becomes disrupted in the diseased state.


Subject(s)
Cardiomyopathy, Hypertrophic, Familial/metabolism , Liver/metabolism , Myocardium/metabolism , Animals , Female , Male
19.
Nat Med ; 19(9): 1153-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933983

ABSTRACT

Preventing reproduction during nutritional deprivation is an adaptive process that is conserved and essential for the survival of species. In mammals, the mechanisms that inhibit fertility during starvation are complex and incompletely understood. Here we show that exposure of female mice to fibroblast growth factor 21 (FGF21), a fasting-induced hepatokine, mimics infertility secondary to starvation. Mechanistically, FGF21 acts on the suprachiasmatic nucleus (SCN) in the hypothalamus to suppress the vasopressin-kisspeptin signaling cascade, thereby inhibiting the proestrus surge in luteinizing hormone. Mice lacking the FGF21 co-receptor, ß-Klotho, in the SCN are refractory to the inhibitory effect of FGF21 on female fertility. Thus, FGF21 defines an important liver-neuroendocrine axis that modulates female reproduction in response to nutritional challenge.


Subject(s)
Fibroblast Growth Factors/metabolism , Infertility, Female/metabolism , Membrane Proteins/metabolism , Reproduction , Starvation/metabolism , Animals , Energy Metabolism , Female , Hypothalamus , Kisspeptins/antagonists & inhibitors , Kisspeptins/metabolism , Klotho Proteins , Luteinizing Hormone/biosynthesis , Luteinizing Hormone/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proestrus/physiology , Signal Transduction , Suprachiasmatic Nucleus , Vasopressins/antagonists & inhibitors , Vasopressins/metabolism
20.
Nat Med ; 19(9): 1147-52, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933984

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a hepatokine that acts as a global starvation signal to modulate fuel partitioning and metabolism and repress growth; however, the site of action of these diverse effects remains unclear. FGF21 signals through a heteromeric cell-surface receptor composed of one of three FGF receptors (FGFR1c, FGFR2c or FGFR3c) in complex with ß-Klotho, a single-pass transmembrane protein that is enriched in metabolic tissues. Here we show that in addition to its known effects on peripheral metabolism, FGF21 increases systemic glucocorticoid levels, suppresses physical activity and alters circadian behavior, which are all features of the adaptive starvation response. These effects are mediated through ß-Klotho expression in the suprachiasmatic nucleus of the hypothalamus and the dorsal vagal complex of the hindbrain. Mice lacking the gene encoding ß-Klotho (Klb) in these regions are refractory to these effects, as well as those on metabolism, insulin and growth. These findings demonstrate a crucial role for the nervous system in mediating the diverse physiologic and pharmacologic actions of FGF21.


Subject(s)
Circadian Rhythm/physiology , Energy Metabolism , Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Nervous System/metabolism , Animals , Glucocorticoids/metabolism , Hypothalamus/metabolism , Klotho Proteins , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Motor Activity , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction , Starvation , Suprachiasmatic Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...