Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Elife ; 122024 May 22.
Article in English | MEDLINE | ID: mdl-38775132

ABSTRACT

The energy-burning capability of beige adipose tissue is a potential therapeutic tool for reducing obesity and metabolic disease, but this capacity is decreased by aging. Here, we evaluate the impact of aging on the profile and activity of adipocyte stem and progenitor cells (ASPCs) and adipocytes during the beiging process in mice. We found that aging increases the expression of Cd9 and other fibro-inflammatory genes in fibroblastic ASPCs and blocks their differentiation into beige adipocytes. Fibroblastic ASPC populations from young and aged mice were equally competent for beige differentiation in vitro, suggesting that environmental factors suppress adipogenesis in vivo. Examination of adipocytes by single nucleus RNA-sequencing identified compositional and transcriptional differences in adipocyte populations with aging and cold exposure. Notably, cold exposure induced an adipocyte population expressing high levels of de novo lipogenesis (DNL) genes, and this response was severely blunted in aged animals. We further identified Npr3, which encodes the natriuretic peptide clearance receptor, as a marker gene for a subset of white adipocytes and an aging-upregulated gene in adipocytes. In summary, this study indicates that aging blocks beige adipogenesis and dysregulates adipocyte responses to cold exposure and provides a resource for identifying cold and aging-regulated pathways in adipose tissue.


Subject(s)
Adipocytes, Beige , Adipogenesis , Aging , Cold Temperature , Animals , Adipogenesis/genetics , Aging/metabolism , Aging/physiology , Mice , Adipocytes, Beige/metabolism , Mice, Inbred C57BL , Male , Adipocytes/metabolism , Cell Differentiation , Cellular Reprogramming , Metabolic Reprogramming
2.
Mol Metab ; 80: 101880, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38218536

ABSTRACT

OBJECTIVE: Glucagon-like peptide 1 (GLP-1) receptor agonists reduce food intake, producing remarkable weight loss in overweight and obese individuals. While much of this weight loss is fat mass, there is also a loss of lean mass, similar to other approaches that induce calorie deficit. Targeting signaling pathways that regulate skeletal muscle hypertrophy is a promising avenue to preserve lean mass and modulate body composition. Myostatin and Activin A are TGFß-like ligands that signal via the activin type II receptors (ActRII) to antagonize muscle growth. Pre-clinical and clinical studies demonstrate that ActRII blockade induces skeletal muscle hypertrophy and reduces fat mass. In this manuscript, we test the hypothesis that combined ActRII blockade and GLP-1 receptor agonism will preserve muscle mass, leading to improvements in skeletomuscular and metabolic function and enhanced fat loss. METHODS: In this study, we explore the therapeutic potential of bimagrumab, a monoclonal antibody against ActRII, to modify body composition alone and during weight loss induced by GLP-1 receptor agonist semaglutide in diet-induced obese mice. Mechanistically, we define the specific role of the anabolic kinase Akt in mediating the hypertrophic muscle effects of ActRII inhibition in vivo. RESULTS: Treatment of obese mice with bimagrumab induced a ∼10 % increase in lean mass while simultaneously decreasing fat mass. Daily treatment of obese mice with semaglutide potently decreased body weight; this included a significant decrease in both muscle and fat mass. Combination treatment with bimagrumab and semaglutide led to superior fat mass loss while simultaneously preserving lean mass despite reduced food intake. Treatment with both drugs was associated with improved metabolic outcomes, and increased lean mass was associated with improved exercise performance. Deletion of both Akt isoforms in skeletal muscle modestly reduced, but did not prevent, muscle hypertrophy driven by ActRII inhibition. CONCLUSIONS: Collectively, these data demonstrate that blockade of ActRII signaling improves body composition and metabolic parameters during calorie deficit driven by GLP-1 receptor agonism and demonstrate the existence of Akt-independent pathways supporting muscle hypertrophy in the absence of ActRII signaling.


Subject(s)
Activin Receptors, Type II , Antibodies, Monoclonal, Humanized , Glucagon-Like Peptide-1 Receptor , Obesity , Proto-Oncogene Proteins c-akt , Weight Loss , Animals , Mice , Activin Receptors, Type II/antagonists & inhibitors , Activin Receptors, Type II/metabolism , Activins/metabolism , Antibodies, Blocking/metabolism , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hypertrophy/metabolism , Mice, Obese , Muscle, Skeletal/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Antibodies, Monoclonal, Humanized/administration & dosage , Obesity/drug therapy
3.
bioRxiv ; 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-36993336

ABSTRACT

The energy-burning capability of beige adipose tissue is a potential therapeutic tool for reducing obesity and metabolic disease, but this capacity is decreased by aging. Here, we evaluate the impact of aging on the profile and activity of adipocyte stem and progenitor cells (ASPCs) and adipocytes during the beiging process. We found that aging increases the expression of Cd9 and other fibro-inflammatory genes in fibroblastic ASPCs and blocks their differentiation into beige adipocytes. Fibroblastic ASPC populations from young and aged mice were equally competent for beige differentiation in vitro, suggesting that environmental factors suppress adipogenesis in vivo. Examination of adipocytes by single nucleus RNA-sequencing identified compositional and transcriptional differences in adipocyte populations with age and cold exposure. Notably, cold exposure induced an adipocyte population expressing high levels of de novo lipogenesis (DNL) genes, and this response was severely blunted in aged animals. We further identified natriuretic peptide clearance receptor Npr3, a beige fat repressor, as a marker gene for a subset of white adipocytes and an aging-upregulated gene in adipocytes. In summary, this study indicates that aging blocks beige adipogenesis and dysregulates adipocyte responses to cold exposure and provides a unique resource for identifying cold and aging-regulated pathways in adipose tissue.

4.
Nat Commun ; 13(1): 4633, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35941104

ABSTRACT

Cancer cachexia is a common, debilitating condition with limited therapeutic options. Using an established mouse model of lung cancer, we find that cachexia is characterized by reduced food intake, spontaneous activity, and energy expenditure accompanied by muscle metabolic dysfunction and atrophy. We identify Activin A as a purported driver of cachexia and treat with ActRIIB-Fc, a decoy ligand for TGF-ß/activin family members, together with anamorelin (Ana), a ghrelin receptor agonist, to reverse muscle dysfunction and anorexia, respectively. Ana effectively increases food intake but only the combination of drugs increases lean mass, restores spontaneous activity, and improves overall survival. These beneficial effects are limited to female mice and are dependent on ovarian function. In agreement, high expression of Activin A in human lung adenocarcinoma correlates with unfavorable prognosis only in female patients, despite similar expression levels in both sexes. This study suggests that multimodal, sex-specific, therapies are needed to reverse cachexia.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Animals , Anorexia/complications , Appetite , Cachexia/drug therapy , Cachexia/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/metabolism , Male , Mice
5.
J Biol Chem ; 297(1): 100815, 2021 07.
Article in English | MEDLINE | ID: mdl-34023388

ABSTRACT

Cyclic AMP-responsive element-binding protein H (CREBH encoded by Creb3l3) is a transcription factor that regulates the expression of genes that control lipid and glucose metabolism as well as inflammation. CREBH is upregulated in the liver under conditions of overnutrition, and mice globally lacking the gene (CREBH-/-) are highly susceptible to diet-induced obesity, insulin resistance, and hepatic steatosis. The net protective effects of CREBH have been attributed in large part to the activities of fibroblast growth factor (Fgf)-21 (Fgf21), a target gene that promotes weight loss, improves glucose homeostasis, and reduces hepatic lipid accumulation. To explore the possibility that activation of the CREBH-Fgf21 axis could ameliorate established effects of high-fat feeding, we generated an inducible transgenic hepatocyte-specific CREBH overexpression mouse model (Tg-rtTA). Acute overexpression of CREBH in livers of Tg-rtTA mice effectively reversed diet-induced obesity, insulin resistance, and hepatic steatosis. These changes were associated with increased activities of thermogenic brown and beige adipose tissues in Tg-rtTA mice, leading to reductions in fat mass, along with enhanced insulin sensitivity and glucose tolerance. Genetically silencing Fgf21 in Tg-rtTA mice abrogated the CREBH-mediated reductions in body weight loss, but only partially reversed the observed improvements in glucose metabolism. These findings reveal that the protective effects of CREBH activation may be leveraged to mitigate diet-induced obesity and associated metabolic abnormalities in both Fgf21-dependent and Fgf21-independent pathways.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Diet , Fatty Liver/genetics , Fatty Liver/pathology , Insulin Resistance/genetics , Liver/metabolism , Obesity/genetics , Adiposity , Animals , Body Weight , Cyclic AMP Response Element-Binding Protein/metabolism , Energy Metabolism , Feeding Behavior , Fibroblast Growth Factors/metabolism , Liver/pathology , Mice, Inbred C57BL , Mice, Knockout
6.
Nat Metab ; 3(4): 469-484, 2021 04.
Article in English | MEDLINE | ID: mdl-33846639

ABSTRACT

Brown adipose tissue can expend large amounts of energy, and therefore increasing its size or activity is a promising therapeutic approach to combat metabolic disease. In humans, major deposits of brown fat cells are found intimately associated with large blood vessels, corresponding to perivascular adipose tissue (PVAT). However, the cellular origins of PVAT are poorly understood. Here, we determine the identity of perivascular adipocyte progenitors in mice and humans. In mice, thoracic PVAT develops from a fibroblastic lineage, consisting of progenitor cells (Pdgfra+, Ly6a+ and Pparg-) and preadipocytes (Pdgfra+, Ly6a+ and Pparg+), which share transcriptional similarity with analogous cell types in white adipose tissue. Interestingly, the aortic adventitia of adult animals contains a population of adipogenic smooth muscle cells (Myh11+, Pdgfra- and Pparg+) that contribute to perivascular adipocyte formation. Similarly, human PVAT contains presumptive fibroblastic and smooth muscle-like adipocyte progenitor cells, as revealed by single-nucleus RNA sequencing. Together, these studies define distinct populations of progenitor cells for thermogenic PVAT, providing a foundation for developing strategies to augment brown fat activity.


Subject(s)
Adipocytes, Brown/physiology , Adipose Tissue, Brown/physiology , Cell Lineage/physiology , Thermogenesis/physiology , Adipocytes, White/physiology , Adipogenesis/physiology , Adipose Tissue, Brown/growth & development , Animals , Animals, Newborn , Aorta/cytology , Aorta/physiology , Blood Vessels/physiology , Cell Lineage/genetics , Fibroblasts/physiology , Gene Expression Regulation/physiology , Humans , Infant, Newborn , Mice , Mice, Inbred C57BL , Myocytes, Smooth Muscle/physiology , Stem Cells/physiology , Thermogenesis/genetics
7.
Hepatology ; 72(3): 857-872, 2020 09.
Article in English | MEDLINE | ID: mdl-32498134

ABSTRACT

BACKGROUND AND AIMS: Obesity-induced pathogenesis of nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH) is associated with increased de novo lipogenesis (DNL) and hepatic glucose production (HGP) that is due to excess fatty acids. Acyl-coenzyme A (CoA) thioesterase (Acot) family members control the cellular utilization of fatty acids by hydrolyzing (deactivating) acyl-CoA into nonesterified fatty acids and CoASH. APPROACH AND RESULTS: Using Caenorhabditis elegans, we identified Acot9 as the strongest regulator of lipid accumulation within the Acot family. Indicative of a maladaptive function, hepatic Acot9 expression was higher in patients with obesity who had NAFLD and NASH compared with healthy controls with obesity. In the setting of excessive nutrition, global ablation of Acot9 protected mice against increases in weight gain, HGP, steatosis, and steatohepatitis. Supportive of a hepatic function, the liver-specific deletion of Acot9 inhibited HGP and steatosis in mice without affecting diet-induced weight gain. By contrast, the rescue of Acot9 expression only in the livers of Acot9 knockout mice was sufficient to promote HGP and steatosis. Mechanistically, hepatic Acot9 localized to the inner mitochondrial membrane, where it deactivated short-chain but not long-chain fatty acyl-CoA. This unique localization and activity of Acot9 directed acetyl-CoA away from protein lysine acetylation and toward the citric acid (TCA) cycle. Acot9-mediated exacerbation of triglyceride and glucose biosynthesis was attributable at least in part to increased TCA cycle activity, which provided substrates for HGP and DNL. ß-oxidation and ketone body production, which depend on long-chain fatty acyl-CoA, were not regulated by Acot9. CONCLUSIONS: Taken together, our findings indicate that Acot9 channels hepatic acyl-CoAs toward increased HGP and DNL under the pathophysiology of obesity. Therefore, Acot9 represents a target for the management of NAFLD.


Subject(s)
Acyl Coenzyme A/metabolism , Fatty Acids/metabolism , Fatty Liver/metabolism , Lipogenesis , Non-alcoholic Fatty Liver Disease/metabolism , Obesity/metabolism , Thiolester Hydrolases , Animals , Caenorhabditis elegans , Drug Discovery , Gene Deletion , Glucose/biosynthesis , Humans , Liver/metabolism , Mice , Mice, Knockout , Thiolester Hydrolases/genetics , Thiolester Hydrolases/metabolism
8.
Cell Metab ; 31(3): 592-604.e9, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32084379

ABSTRACT

Recent studies suggest that a key mechanism whereby the gut microbiome influences energy balance and glucose homeostasis is through the recruitment of brown and beige adipocytes, primary mediators of the adaptive thermogenic response. To test this, we assessed energy expenditure and glucose metabolism in two complementary mouse models of gut microbial deficiency, which were exposed to a broad range of thermal and dietary stresses. Neither ablation of the gut microbiome, nor the substantial microbial perturbations induced by cold ambient temperatures, influenced energy expenditure during cold exposure or high-fat feeding. Nevertheless, we demonstrated a critical role for gut microbial metabolism in maintaining euglycemia through the production of amino acid metabolites that optimized hepatic TCA (tricarboxylic acid) cycle fluxes in support of gluconeogenesis. These results distinguish the dispensability of the gut microbiome for the regulation of energy expenditure from its critical contribution to the maintenance of glucose homeostasis.


Subject(s)
Gastrointestinal Microbiome , Glucose/metabolism , Homeostasis , Thermogenesis/physiology , Animals , Cold Temperature , Diet , Gluconeogenesis , Liver/metabolism , Male , Mice, Inbred C57BL
9.
FASEB J ; 33(10): 11579-11594, 2019 10.
Article in English | MEDLINE | ID: mdl-31339804

ABSTRACT

Fibroblast growth factor (FGF)13, a nonsecreted, X-linked, FGF homologous factor, is differentially expressed in adipocytes in response to diet, yet Fgf13's role in metabolism has not been explored. Heterozygous Fgf13 knockouts fed normal chow and housed at 22°C showed hyperactivity accompanying reduced core temperature and obesity when housed at 30°C. Those heterozygous knockouts showed defects in thermogenesis even at 30°C and an inability to protect core temperature. Surprisingly, we detected trivial FGF13 in adipose of wild-type mice fed normal chow and no obesity in adipose-specific heterozygous knockouts housed at 30°C, and we detected an intact brown fat response through exogenous ß3 agonist stimulation, suggesting a defect in sympathetic drive to brown adipose tissue. In contrast, hypothalamic-specific ablation of Fgf13 recapitulated weight gain at 30°C. Norepinephrine turnover in brown fat was reduced at both housing temperatures. Thus, our data suggest that impaired CNS regulation of sympathetic activation of brown fat underlies obesity and thermogenesis in Fgf13 heterozygous knockouts fed normal chow.-Sinden, D. S., Holman, C. D., Bare, C. J., Sun, X., Gade, A. R., Cohen, D. E., Pitt, G. S. Knockout of the X-linked Fgf13 in the hypothalamic paraventricular nucleus impairs sympathetic output to brown fat and causes obesity.


Subject(s)
Adipose Tissue, Brown/metabolism , Fibroblast Growth Factors/metabolism , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Adipocytes/metabolism , Adipose Tissue, White/metabolism , Adiposity/physiology , Animals , Diet, High-Fat/methods , Energy Metabolism/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Thermogenesis/physiology , Weight Gain/physiology
10.
Hepatology ; 68(6): 2167-2181, 2018 12.
Article in English | MEDLINE | ID: mdl-29698569

ABSTRACT

Integrated stress response (ISR) is a signaling system in which phosphorylation of eukaryotic translation initiation factor 2α (eIF2α) by stress-specific kinases and subsequent activation of activation transcription factor (ATF) 4 help restore cellular homeostasis following exposure to environmental stresses. ISR activation has been observed in metabolic diseases, including hepatic steatosis (HS), steatohepatitis (SH), and insulin resistance (IR), but it remains unclear whether ISR contributes to disease pathogenesis or represents an innate defense mechanism against metabolic stresses. Constitutive repressor of eIF2α phosphorylation (CReP) is a critical regulatory subunit of the eIF2α phosphatase complex. Here, we show that CReP ablation causes constitutive eIF2α phosphorylation in the liver, which leads to activation of the ATF4 transcriptional program including increased fibroblast growth factor 21 (FGF21) production. Liver-specific CReP knockout (CRePLKO ) mice exhibited marked browning of white adipose tissue (WAT) and increased energy expenditure and insulin sensitivity in an FGF21-dependent manner. Furthermore, CRePLKO mice were protected from high-fat diet (HFD)-induced obesity, HS, and IR. Acute CReP ablation in liver of HFD-induced obese mice also reduced adiposity and improved glucose homeostasis. Conclusion: These data suggest that CReP abundance is a critical determinant for eIF2α phosphorylation and ensuing ISR activation in the liver. Constitutive ISR activation in the liver induces FGF21 and confers protection from HFD-induced adiposity, IR, and HS in mice. Augmenting hepatic ISR may represent a therapeutic approach to treat metabolic disorders.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Fatty Liver/etiology , Fibroblast Growth Factors/metabolism , Protein Phosphatase 1/physiology , Stress, Physiological , Activating Transcription Factor 4/metabolism , Adipocytes, Beige/physiology , Adiposity , Animals , Diet, High-Fat/adverse effects , Energy Metabolism , Homeostasis , Insulin Resistance , Mice , Mice, Knockout , Obesity/etiology
11.
Cell Host Microbe ; 23(4): 447-457.e4, 2018 04 11.
Article in English | MEDLINE | ID: mdl-29576480

ABSTRACT

Bone marrow transplantation (BMT) offers curative potential for patients with high-risk hematologic malignancies, but the post-transplantation period is characterized by profound immunodeficiency. Recent studies indicate that the intestinal microbiota not only regulates mucosal immunity, but can also contribute to systemic immunity and hematopoiesis. Using antibiotic-mediated microbiota depletion in a syngeneic BMT mouse model, here we describe a role for the intestinal flora in hematopoietic recovery after BMT. Depletion of the intestinal microbiota resulted in impaired recovery of lymphocyte and neutrophil counts, while recovery of the hematopoietic stem and progenitor compartments and the erythroid lineage were largely unaffected. Depletion of the intestinal microbiota also reduced dietary energy uptake and visceral fat stores. Caloric supplementation through sucrose in the drinking water improved post-BMT hematopoietic recovery in mice with a depleted intestinal flora. Taken together, we show that the intestinal microbiota contribute to post-BMT hematopoietic reconstitution in mice through improved dietary energy uptake.


Subject(s)
Bone Marrow Transplantation , Gastrointestinal Microbiome , Nutritional Support , Animals , Bone Marrow/physiology , Hematopoiesis , Mice , Models, Animal , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...