Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
1.
Cell ; 182(2): 372-387.e14, 2020 07 23.
Article in English | MEDLINE | ID: mdl-32610084

ABSTRACT

Acute psychological stress has long been known to decrease host fitness to inflammation in a wide variety of diseases, but how this occurs is incompletely understood. Using mouse models, we show that interleukin-6 (IL-6) is the dominant cytokine inducible upon acute stress alone. Stress-inducible IL-6 is produced from brown adipocytes in a beta-3-adrenergic-receptor-dependent fashion. During stress, endocrine IL-6 is the required instructive signal for mediating hyperglycemia through hepatic gluconeogenesis, which is necessary for anticipating and fueling "fight or flight" responses. This adaptation comes at the cost of enhancing mortality to a subsequent inflammatory challenge. These findings provide a mechanistic understanding of the ontogeny and adaptive purpose of IL-6 as a bona fide stress hormone coordinating systemic immunometabolic reprogramming. This brain-brown fat-liver axis might provide new insights into brown adipose tissue as a stress-responsive endocrine organ and mechanistic insight into targeting this axis in the treatment of inflammatory and neuropsychiatric diseases.


Subject(s)
Adipose Tissue, Brown/metabolism , Interleukin-6/metabolism , Stress, Psychological , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Brain/metabolism , Chemokines/metabolism , Cytokines/metabolism , Disease Models, Animal , Gluconeogenesis , Hyperglycemia/metabolism , Hyperglycemia/pathology , Interleukin-6/blood , Interleukin-6/genetics , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-3/metabolism , Receptors, Interleukin-6/metabolism , Uncoupling Protein 1/deficiency , Uncoupling Protein 1/genetics
2.
Cell ; 183(3): 684-701.e14, 2020 10 29.
Article in English | MEDLINE | ID: mdl-33058756

ABSTRACT

Positive selection in Europeans at the 2q21.3 locus harboring the lactase gene has been attributed to selection for the ability of adults to digest milk to survive famine in ancient times. However, the 2q21.3 locus is also associated with obesity and type 2 diabetes in humans, raising the possibility that additional genetic elements in the locus may have contributed to evolutionary adaptation to famine by promoting energy storage, but which now confer susceptibility to metabolic diseases. We show here that the miR-128-1 microRNA, located at the center of the positively selected locus, represents a crucial metabolic regulator in mammals. Antisense targeting and genetic ablation of miR-128-1 in mouse metabolic disease models result in increased energy expenditure and amelioration of high-fat-diet-induced obesity and markedly improved glucose tolerance. A thrifty phenotype connected to miR-128-1-dependent energy storage may link ancient adaptation to famine and modern metabolic maladaptation associated with nutritional overabundance.


Subject(s)
Metabolic Diseases/genetics , MicroRNAs/genetics , Adipocytes, Brown/pathology , Adiposity , Alleles , Animals , Cell Differentiation , Cell Line , Cells, Cultured , Diet, High-Fat , Energy Metabolism , Epigenesis, Genetic , Genetic Loci , Glucose/metabolism , Homeostasis , Humans , Hypertrophy , Insulin Resistance , Leptin/deficiency , Leptin/metabolism , Male , Mammals/genetics , Mice, Inbred C57BL , Mice, Obese , MicroRNAs/metabolism , Obesity/genetics , Oligonucleotides/metabolism , Species Specificity
3.
Cell ; 172(1-2): 234-248.e17, 2018 01 11.
Article in English | MEDLINE | ID: mdl-29307489

ABSTRACT

The transition from the fed to the fasted state necessitates a shift from carbohydrate to fat metabolism that is thought to be mostly orchestrated by reductions in plasma insulin concentrations. Here, we show in awake rats that insulinopenia per se does not cause this transition but that both hypoleptinemia and insulinopenia are necessary. Furthermore, we show that hypoleptinemia mediates a glucose-fatty acid cycle through activation of the hypothalamic-pituitary-adrenal axis, resulting in increased white adipose tissue (WAT) lipolysis rates and increased hepatic acetyl-coenzyme A (CoA) content, which are essential to maintain gluconeogenesis during starvation. We also show that in prolonged starvation, substrate limitation due to reduced rates of glucose-alanine cycling lowers rates of hepatic mitochondrial anaplerosis, oxidation, and gluconeogenesis. Taken together, these data identify a leptin-mediated glucose-fatty acid cycle that integrates responses of the muscle, WAT, and liver to promote a shift from carbohydrate to fat oxidation and maintain glucose homeostasis during starvation.


Subject(s)
Blood Glucose/metabolism , Fatty Acids/metabolism , Gluconeogenesis , Homeostasis , Leptin/metabolism , Starvation/metabolism , Adipose Tissue, White/metabolism , Alanine/metabolism , Animals , Insulin/blood , Leptin/blood , Lipolysis , Liver/metabolism , Male , Mitochondria/metabolism , Rats , Rats, Sprague-Dawley
4.
Cell ; 169(1): 148-160.e15, 2017 03 23.
Article in English | MEDLINE | ID: mdl-28340340

ABSTRACT

Type 2 diabetes (T2D) is a worldwide epidemic with a medical need for additional targeted therapies. Suppression of hepatic glucose production (HGP) effectively ameliorates diabetes and can be exploited for its treatment. We hypothesized that targeting PGC-1α acetylation in the liver, a chemical modification known to inhibit hepatic gluconeogenesis, could be potentially used for treatment of T2D. Thus, we designed a high-throughput chemical screen platform to quantify PGC-1α acetylation in cells and identified small molecules that increase PGC-1α acetylation, suppress gluconeogenic gene expression, and reduce glucose production in hepatocytes. On the basis of potency and bioavailability, we selected a small molecule, SR-18292, that reduces blood glucose, strongly increases hepatic insulin sensitivity, and improves glucose homeostasis in dietary and genetic mouse models of T2D. These studies have important implications for understanding the regulatory mechanisms of glucose metabolism and treatment of T2D.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Gluconeogenesis/drug effects , Hypoglycemic Agents/administration & dosage , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors , Acetylation , Animals , Blood Glucose/metabolism , Cells, Cultured , Glucose/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Hepatocytes/metabolism , High-Throughput Screening Assays , Insulin Resistance , Mice , p300-CBP Transcription Factors/metabolism
6.
Cell ; 160(4): 745-758, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25662011

ABSTRACT

Impaired insulin-mediated suppression of hepatic glucose production (HGP) plays a major role in the pathogenesis of type 2 diabetes (T2D), yet the molecular mechanism by which this occurs remains unknown. Using a novel in vivo metabolomics approach, we show that the major mechanism by which insulin suppresses HGP is through reductions in hepatic acetyl CoA by suppression of lipolysis in white adipose tissue (WAT) leading to reductions in pyruvate carboxylase flux. This mechanism was confirmed in mice and rats with genetic ablation of insulin signaling and mice lacking adipose triglyceride lipase. Insulin's ability to suppress hepatic acetyl CoA, PC activity, and lipolysis was lost in high-fat-fed rats, a phenomenon reversible by IL-6 neutralization and inducible by IL-6 infusion. Taken together, these data identify WAT-derived hepatic acetyl CoA as the main regulator of HGP by insulin and link it to inflammation-induced hepatic insulin resistance associated with obesity and T2D.


Subject(s)
Acetyl Coenzyme A/metabolism , Insulin Resistance , Liver/metabolism , Panniculitis/metabolism , Adipose Tissue, White/chemistry , Adolescent , Animals , Diabetes Mellitus, Type 2 , Diet, High-Fat , Glucose/metabolism , Humans , Hyperglycemia , Interleukin-6/analysis , Lipolysis , Male , Mice , Obesity/metabolism , Rats, Sprague-Dawley
7.
Genes Dev ; 35(1-2): 133-146, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33334822

ABSTRACT

The cJun NH2-terminal kinase (JNK) signaling pathway is activated by metabolic stress and promotes the development of metabolic syndrome, including hyperglycemia, hyperlipidemia, and insulin resistance. This integrated physiological response involves cross-talk between different organs. Here we demonstrate that JNK signaling in adipocytes causes an increased circulating concentration of the hepatokine fibroblast growth factor 21 (FGF21) that regulates systemic metabolism. The mechanism of organ crosstalk is mediated by a feed-forward regulatory loop caused by JNK-regulated FGF21 autocrine signaling in adipocytes that promotes increased expression of the adipokine adiponectin and subsequent hepatic expression of the hormone FGF21. The mechanism of organ cross-talk places circulating adiponectin downstream of autocrine FGF21 expressed by adipocytes and upstream of endocrine FGF21 expressed by hepatocytes. This regulatory loop represents a novel signaling paradigm that connects autocrine and endocrine signaling modes of the same hormone in different tissues.


Subject(s)
Adipose Tissue/physiology , Autocrine Communication/genetics , Fibroblast Growth Factors/genetics , Gene Expression Regulation/genetics , Signal Transduction/genetics , Adipocytes/metabolism , Adiponectin/metabolism , Adipose Tissue/physiopathology , Animals , Endocrine System/metabolism , Energy Metabolism/genetics , Feedback, Physiological/physiology , Fibroblast Growth Factors/blood , Hepatocytes/metabolism , Insulin Resistance/genetics , Liver/metabolism , MAP Kinase Kinase 4/deficiency , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , MAP Kinase Signaling System/physiology , Mice
8.
Nature ; 600(7888): 314-318, 2021 12.
Article in English | MEDLINE | ID: mdl-34819664

ABSTRACT

Thermogenesis in brown and beige adipose tissue has important roles in maintaining body temperature and countering the development of metabolic disorders such as obesity and type 2 diabetes1,2. Although much is known about commitment and activation of brown and beige adipose tissue, its multiple and abundant immunological factors have not been well characterized3-6. Here we define a critical role of IL-27-IL-27Rα signalling in improving thermogenesis, protecting against diet-induced obesity and ameliorating insulin resistance. Mechanistic studies demonstrate that IL-27 directly targets adipocytes, activating p38 MAPK-PGC-1α signalling and stimulating the production of UCP1. Notably, therapeutic administration of IL-27 ameliorated metabolic morbidities in well-established mouse models of obesity. Consistently, individuals with obesity show significantly decreased levels of serum IL-27, which can be restored after bariatric surgery. Collectively, these findings show that IL-27 has an important role in orchestrating metabolic programs, and is a highly promising target for anti-obesity immunotherapy.


Subject(s)
Adipocytes/metabolism , Energy Metabolism , Interleukin-27/metabolism , Thermogenesis , Animals , Bariatric Surgery , Disease Models, Animal , Female , Humans , Insulin Resistance , Interleukin-27/blood , Interleukin-27/therapeutic use , Male , Mice , Obesity/blood , Obesity/drug therapy , Obesity/metabolism , Obesity/prevention & control , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Receptors, Interleukin/metabolism , Signal Transduction , Uncoupling Protein 1/biosynthesis , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Nature ; 579(7798): 279-283, 2020 03.
Article in English | MEDLINE | ID: mdl-32132708

ABSTRACT

Although it is well-established that reductions in the ratio of insulin to glucagon in the portal vein have a major role in the dysregulation of hepatic glucose metabolism in type-2 diabetes1-3, the mechanisms by which glucagon affects hepatic glucose production and mitochondrial oxidation are poorly understood. Here we show that glucagon stimulates hepatic gluconeogenesis by increasing the activity of hepatic adipose triglyceride lipase, intrahepatic lipolysis, hepatic acetyl-CoA content and pyruvate carboxylase flux, while also increasing mitochondrial fat oxidation-all of which are mediated by stimulation of the inositol triphosphate receptor 1 (INSP3R1). In rats and mice, chronic physiological increases in plasma glucagon concentrations increased mitochondrial oxidation of fat in the liver and reversed diet-induced hepatic steatosis and insulin resistance. However, these effects of chronic glucagon treatment-reversing hepatic steatosis and glucose intolerance-were abrogated in Insp3r1 (also known as Itpr1)-knockout mice. These results provide insights into glucagon biology and suggest that INSP3R1 may represent a target for therapies that aim to reverse nonalcoholic fatty liver disease and type-2 diabetes.


Subject(s)
Glucagon/pharmacology , Gluconeogenesis/drug effects , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Liver/drug effects , Acetyl Coenzyme A/metabolism , Adipose Tissue/drug effects , Animals , Diabetes Mellitus, Type 2/physiopathology , Enzyme Activation/drug effects , Glucagon/blood , Inositol 1,4,5-Trisphosphate Receptors/genetics , Lipase/metabolism , Lipolysis/drug effects , Lipolysis/genetics , Mice, Knockout , Mitochondria/drug effects , Non-alcoholic Fatty Liver Disease/physiopathology , Oxidation-Reduction/drug effects
10.
Am J Physiol Endocrinol Metab ; 326(3): E290-E307, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38294698

ABSTRACT

Cancer-related fatigue (CRF) is one of the most prevalent and detrimental complications of cancer. Emerging evidence suggests that obesity and insulin resistance are associated with CRF occurrence and severity in cancer patients and survivors. In this narrative review, we analyzed recent studies including both preclinical and clinical research on the relationship between obesity and/or insulin resistance and CRF. We also describe potential mechanisms for these relationships, though with the caveat that because the mechanisms underlying CRF are incompletely understood, the mechanisms mediating the association between obesity/insulin resistance and CRF are similarly incompletely delineated. The data suggest that, in addition to their effects to worsen CRF by directly promoting tumor growth and metastasis, obesity and insulin resistance may also contribute to CRF by inducing chronic inflammation, neuroendocrinological disturbance, and metabolic alterations. Furthermore, studies suggest that patients with obesity and insulin resistance experience more cancer-induced pain and are at more risk of emotional and behavioral disruptions correlated with CRF. However, other studies implied a potentially paradoxical impact of obesity and insulin resistance to reduce CRF symptoms. Despite the need for further investigation utilizing interventions to directly elucidate the mechanisms of cancer-related fatigue, current evidence demonstrates a correlation between obesity and/or insulin resistance and CRF, and suggests potential therapeutics for CRF by targeting obesity and/or obesity-related mediators.


Subject(s)
Insulin Resistance , Neoplasms , Humans , Neoplasms/complications , Obesity/complications , Fatigue/etiology
11.
Am J Physiol Endocrinol Metab ; 326(3): E341-E350, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38294697

ABSTRACT

Several clinical studies observed a surprising beneficial effect of obesity on enhancing immunotherapy responsiveness in patients with melanoma, highlighting an as-yet insufficiently understood relationship between metabolism and immunogenicity. Here, we demonstrate that the thiazolidinedione (TZD) rosiglitazone, a drug commonly used to treat diabetes by sequestering fatty acids in metabolically inert subcutaneous adipose tissue, improved sensitivity to anti-programmed cell death protein 1 (PD-1) treatment in YUMMER1.7 tumor-bearing mice, an initially immunotherapy-sensitive murine melanoma model. We observed a transition from high to intermediate PD-1 expression in tumor-infiltrating CD8+ T cells. Moreover, TZD inhibited PD-1 expression in mouse and human T cells treated in vitro. In addition to its direct impact on immune cells, TZD also decreased circulating insulin concentrations, while insulin induced T cell exhaustion in culture. In TZD-treated mice, we observed higher fatty acid concentrations in the tumor microenvironment, with fatty acids protecting against exhaustion in culture. Together, these data are consistent with an indirect mechanism of TZD inhibiting T cell exhaustion. Finally, we analyzed imaging data from patients with melanoma before and after anti-PD-1 treatment, confirming the beneficial effect of increased subcutaneous fat on anti-PD-1 responsiveness in patients. We also found that the expression of peroxisome proliferator-activated receptor gamma (PPARγ), the canonical activator of lipid uptake and adipogenesis activated by TZD, correlated with overall survival time. Taken together, these data identify a new adjuvant to enhance immunotherapy efficacy in YUMMER1.7 melanoma mice, and discover a new metabolism-based prognostic marker in human melanoma.NEW & NOTEWORTHY Zhang et al. demonstrate that the diabetes drug rosiglitazone improves the efficacy of immunotherapy in mouse melanoma. This effect is both direct and indirect: TZD directly reduces PD-1 expression in CD8+ T cells (i.e., reduces exhaustion), and indirectly reduces exhaustion by lowering insulin levels and increasing local fat. Finally, they demonstrate that hallmarks of TZD action (such as PPARγ expression and subcutaneous fat content) correlate with improved immunotherapy efficacy in humans with melanoma.


Subject(s)
Diabetes Mellitus , Melanoma , Thiazolidinediones , Humans , Animals , Mice , Melanoma/drug therapy , Rosiglitazone , Programmed Cell Death 1 Receptor , PPAR gamma , Thiazolidinediones/pharmacology , Thiazolidinediones/therapeutic use , Antibodies, Monoclonal , Insulin , Fatty Acids , Tumor Microenvironment
12.
Diabetologia ; 66(3): 567-578, 2023 03.
Article in English | MEDLINE | ID: mdl-36456864

ABSTRACT

AIMS/HYPOTHESIS: Athletes exhibit increased muscle insulin sensitivity, despite increased intramuscular triacylglycerol content. This phenomenon has been coined the 'athlete's paradox' and is poorly understood. Recent findings suggest that the subcellular distribution of sn-1,2-diacylglycerols (DAGs) in the plasma membrane leading to activation of novel protein kinase Cs (PKCs) is a crucial pathway to inducing insulin resistance. Here, we hypothesised that regular aerobic exercise would preserve muscle insulin sensitivity by preventing increases in plasma membrane sn-1,2-DAGs and activation of PKCε and PKCθ despite promoting increases in muscle triacylglycerol content. METHODS: C57BL/6J mice were allocated to three groups (regular chow feeding [RC]; high-fat diet feeding [HFD]; RC feeding and running wheel exercise [RC-EXE]). We used a novel LC-MS/MS/cellular fractionation method to assess DAG stereoisomers in five subcellular compartments (plasma membrane [PM], endoplasmic reticulum, mitochondria, lipid droplets and cytosol) in the skeletal muscle. RESULTS: We found that the HFD group had a greater content of sn-DAGs and ceramides in multiple subcellular compartments compared with the RC mice, which was associated with an increase in PKCε and PKCθ translocation. However, the RC-EXE mice showed, of particular note, a reduction in PM sn-1,2-DAG and ceramide content when compared with HFD mice. Consistent with the PM sn-1,2-DAG-novel PKC hypothesis, we observed an increase in phosphorylation of threonine1150 on the insulin receptor kinase (IRKT1150), and reductions in insulin-stimulated IRKY1162 phosphorylation and IRS-1-associated phosphoinositide 3-kinase activity in HFD compared with RC and RC-EXE mice, which are sites of PKCε and PKCθ action, respectively. CONCLUSIONS/INTERPRETATION: These results demonstrate that lower PKCθ/PKCε activity and sn-1,2-DAG content, especially in the PM compartment, can explain the preserved muscle insulin sensitivity in RC-EXE mice.


Subject(s)
Insulin Resistance , Mice , Animals , Insulin Resistance/physiology , Protein Kinase C-theta/metabolism , Protein Kinase C-epsilon/metabolism , Chromatography, Liquid , Phosphatidylinositol 3-Kinases/metabolism , Mice, Inbred C57BL , Tandem Mass Spectrometry , Insulin/metabolism , Muscle, Skeletal/metabolism , Triglycerides/metabolism , Ceramides/metabolism
13.
Am J Physiol Endocrinol Metab ; 325(4): E363-E375, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37646579

ABSTRACT

Cancer-related fatigue (CRF) is one of the most common complications in patients with multiple cancer types and severely affects patients' quality of life. However, there have only been single symptom-relieving adjuvant therapies but no effective pharmaceutical treatment for the CRF syndrome. Dichloroacetate (DCA), a small molecule inhibitor of pyruvate dehydrogenase kinase, has been tested as a potential therapy to slow tumor growth, based largely on its effects in vitro to halt cell division. We found that although DCA did not affect rates of tumor growth or the efficacy of standard cancer treatment (immunotherapy and chemotherapy) in two murine cancer models, DCA preserved physical function in mice with late-stage tumors by reducing circulating lactate concentrations. In vivo liquid chromatography-mass spectrometry/mass spectrometry studies suggest that DCA treatment may preserve membrane potential, postpone proteolysis, and relieve oxidative stress in muscles of tumor-bearing mice. In all, this study provides evidence for DCA as a novel pharmaceutical treatment to maintain physical function and motivation in murine models of CRF.NEW & NOTEWORTHY We identify a new metabolic target for cancer-related fatigue, dichloroacetate (DCA). They demonstrate that in mice, DCA preserves physical function and protects against the detrimental effects of cancer treatment by reducing cancer-induced increases in circulating lactate. As DCA is already FDA approved for another indication, these results could be rapidly translated to clinical trials for this condition for which no pharmaceutical therapies exist beyond symptom management.


Subject(s)
Dichloroacetic Acid , Fatigue , Melanoma , Quality of Life , Animals , Mice , Dichloroacetic Acid/pharmacology , Dichloroacetic Acid/therapeutic use , Fatigue/drug therapy , Fatigue/etiology , Lactic Acid/metabolism , Melanoma/complications
14.
Am J Physiol Endocrinol Metab ; 325(1): E46-E61, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37224467

ABSTRACT

Adipose tissues accumulate excess energy as fat and heavily influence metabolic homeostasis. O-linked N-acetylglucosamine (O-GlcNAc) modification (O-GlcNAcylation), which involves the addition of N-acetylglucosamine to proteins by O-GlcNAc transferase (Ogt), modulates multiple cellular processes. However, little is known about the role of O-GlcNAcylation in adipose tissues during body weight gain due to overnutrition. Here, we report on O-GlcNAcylation in mice with high-fat diet (HFD)-induced obesity. Mice with knockout of Ogt in adipose tissue achieved using adiponectin promoter-driven Cre recombinase (Ogt-FKO) gained less body weight than control mice under HFD. Surprisingly, Ogt-FKO mice exhibited glucose intolerance and insulin resistance, despite their reduced body weight gain, as well as decreased expression of de novo lipogenesis genes and increased expression of inflammatory genes, resulting in fibrosis at 24 weeks of age. Primary cultured adipocytes derived from Ogt-FKO mice showed decreased lipid accumulation. Both primary cultured adipocytes and 3T3-L1 adipocytes treated with OGT inhibitor showed increased secretion of free fatty acids. Medium derived from these adipocytes stimulated inflammatory genes in RAW 264.7 macrophages, suggesting that cell-to-cell communication via free fatty acids might be a cause of adipose inflammation in Ogt-FKO mice. In conclusion, O-GlcNAcylation is important for healthy adipose expansion in mice. Glucose flux into adipose tissues may be a signal to store excess energy as fat.NEW & NOTEWORTHY We evaluated the role of O-GlcNAcylation in adipose tissue in diet-induced obesity using adipose tissue-specific Ogt knockout mice. We found that O-GlcNAcylation in adipose tissue is essential for healthy fat expansion and that Ogt-FKO mice exhibit severe fibrosis upon long-term overnutrition. O-GlcNAcylation in adipose tissue may regulate de novo lipogenesis and free fatty acid efflux to the degree of overnutrition. We believe that these results provide new insights into adipose tissue physiology and obesity research.


Subject(s)
Acetylglucosamine , Fatty Acids, Nonesterified , Animals , Mice , Acetylglucosamine/metabolism , Obesity/genetics , Obesity/metabolism , Adipose Tissue/metabolism , Body Weight/genetics , Weight Gain , N-Acetylglucosaminyltransferases/genetics , N-Acetylglucosaminyltransferases/metabolism
15.
Biochem J ; 479(5): 583-607, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35244142

ABSTRACT

For a century, since the pioneering work of Otto Warburg, the interwoven relationship between metabolism and cancer has been appreciated. More recently, with obesity rates rising in the U.S. and worldwide, epidemiologic evidence has supported a link between obesity and cancer. A substantial body of work seeks to mechanistically unpack the association between obesity, altered metabolism, and cancer. Without question, these relationships are multifactorial and cannot be distilled to a single obesity- and metabolism-altering hormone, substrate, or factor. However, it is important to understand the hormone-specific associations between metabolism and cancer. Here, we review the links between obesity, metabolic dysregulation, insulin, and cancer, with an emphasis on current investigational metabolic adjuncts to standard-of-care cancer treatment.


Subject(s)
Insulin , Neoplasms , Humans , Neoplasms/epidemiology , Obesity
16.
Proc Natl Acad Sci U S A ; 117(51): 32584-32593, 2020 12 22.
Article in English | MEDLINE | ID: mdl-33293421

ABSTRACT

Adiponectin has emerged as a potential therapy for type 2 diabetes mellitus, but the molecular mechanism by which adiponectin reverses insulin resistance remains unclear. Two weeks of globular adiponectin (gAcrp30) treatment reduced fasting plasma glucose, triglyceride (TAG), and insulin concentrations and reversed whole-body insulin resistance, which could be attributed to both improved insulin-mediated suppression of endogenous glucose production and increased insulin-stimulated glucose uptake in muscle and adipose tissues. These improvements in liver and muscle sensitivity were associated with ∼50% reductions in liver and muscle TAG and plasma membrane (PM)-associated diacylglycerol (DAG) content and occurred independent of reductions in total ceramide content. Reductions of PM DAG content in liver and skeletal muscle were associated with reduced PKCε translocation in liver and reduced PKCθ and PKCε translocation in skeletal muscle resulting in increased insulin-stimulated insulin receptor tyrosine1162 phosphorylation, IRS-1/IRS-2-associated PI3-kinase activity, and Akt-serine phosphorylation. Both gAcrp30 and full-length adiponectin (Acrp30) treatment increased eNOS/AMPK activation in muscle and muscle fatty acid oxidation. gAcrp30 and Acrp30 infusions also increased TAG uptake in epididymal white adipose tissue (eWAT), which could be attributed to increased lipoprotein lipase (LPL) activity. These data suggest that adiponectin and adiponectin-related molecules reverse lipid-induced liver and muscle insulin resistance by reducing ectopic lipid storage in these organs, resulting in decreased plasma membrane sn-1,2-DAG-induced nPKC activity and increased insulin signaling. Adiponectin mediates these effects by both promoting the storage of TAG in eWAT likely through stimulation of LPL as well as by stimulation of AMPK in muscle resulting in increased muscle fat oxidation.


Subject(s)
Adiponectin/pharmacology , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , AMP-Activated Protein Kinases/metabolism , Adiponectin/genetics , Adiponectin/metabolism , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Animals , Diglycerides/metabolism , Insulin/metabolism , Lipid Metabolism , Lipoprotein Lipase/metabolism , Liver/metabolism , Male , Mice, Inbred C57BL , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Nitric Oxide Synthase Type III/metabolism , Protein Kinase C/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology
17.
Proc Natl Acad Sci U S A ; 117(6): 2751-2760, 2020 02 11.
Article in English | MEDLINE | ID: mdl-31980524

ABSTRACT

Obesity is associated with a chronic state of low-grade inflammation and progressive tissue infiltration by immune cells and increased expression of inflammatory cytokines. It is established that interleukin 6 (IL6) regulates multiple aspects of metabolism, including glucose disposal, lipolysis, oxidative metabolism, and energy expenditure. IL6 is secreted by many tissues, but the role of individual cell types is unclear. We tested the role of specific cells using a mouse model with conditional expression of the Il6 gene. We found that IL6 derived from adipocytes increased, while IL6 derived from myeloid cells and muscle suppressed, macrophage infiltration of adipose tissue. These opposite actions were associated with a switch of IL6 signaling from a canonical mode (myeloid cells) to a noncanonical trans-signaling mode (adipocytes and muscle) with increased expression of the ADAM10/17 metalloprotease that promotes trans-signaling by the soluble IL6 receptor α. Collectively, these data demonstrate that the source of IL6 production plays a major role in the physiological regulation of metabolism.


Subject(s)
Adipose Tissue/immunology , Interleukin-6/immunology , Obesity/immunology , ADAM10 Protein/genetics , ADAM10 Protein/immunology , ADAM17 Protein/genetics , ADAM17 Protein/immunology , Adipocytes/immunology , Animals , Female , Humans , Interleukin-6/genetics , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Muscle Cells/immunology , Myeloid Cells/immunology , Obesity/genetics , Species Specificity
18.
Proc Natl Acad Sci U S A ; 117(14): 8166-8176, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32188779

ABSTRACT

Multiple insulin-regulated enzymes participate in hepatic glycogen synthesis, and the rate-controlling step responsible for insulin stimulation of glycogen synthesis is unknown. We demonstrate that glucokinase (GCK)-mediated glucose phosphorylation is the rate-controlling step in insulin-stimulated hepatic glycogen synthesis in vivo, by use of the somatostatin pancreatic clamp technique using [13C6]glucose with metabolic control analysis (MCA) in three rat models: 1) regular chow (RC)-fed male rats (control), 2) high fat diet (HFD)-fed rats, and 3) RC-fed rats with portal vein glucose delivery at a glucose infusion rate matched to the control. During hyperinsulinemia, hyperglycemia dose-dependently increased hepatic glycogen synthesis. At similar levels of hyperinsulinemia and hyperglycemia, HFD-fed rats exhibited a decrease and portal delivery rats exhibited an increase in hepatic glycogen synthesis via the direct pathway compared with controls. However, the strong correlation between liver glucose-6-phosphate concentration and net hepatic glycogen synthetic rate was nearly identical in these three groups, suggesting that the main difference between models is the activation of GCK. MCA yielded a high control coefficient for GCK in all three groups. We confirmed these findings in studies of hepatic GCK knockdown using an antisense oligonucleotide. Reduced liver glycogen synthesis in lipid-induced hepatic insulin resistance and increased glycogen synthesis during portal glucose infusion were explained by concordant changes in translocation of GCK. Taken together, these data indicate that the rate of insulin-stimulated hepatic glycogen synthesis is controlled chiefly through GCK translocation.


Subject(s)
Fatty Liver/pathology , Glucokinase/metabolism , Glucose/metabolism , Liver Glycogen/biosynthesis , Liver/metabolism , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Fatty Liver/etiology , Gene Knockdown Techniques , Glucokinase/genetics , Glucose/administration & dosage , Glucose-6-Phosphate/analysis , Glucose-6-Phosphate/metabolism , Humans , Hyperglycemia/etiology , Hyperglycemia/pathology , Hyperinsulinism/etiology , Hyperinsulinism/pathology , Insulin/metabolism , Insulin Resistance , Liver/pathology , Male , Metabolomics , Phosphorylation , Rats
19.
Proc Natl Acad Sci U S A ; 117(28): 16616-16625, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32601203

ABSTRACT

Enhanced inflammation is believed to contribute to overnutrition-induced metabolic disturbance. Nutrient flux has also been shown to be essential for immune cell activation. Here, we report an unexpected role of nutrient-sensing O-linked ß-N-acetylglucosamine (O-GlcNAc) signaling in suppressing macrophage proinflammatory activation and preventing diet-induced metabolic dysfunction. Overnutrition stimulates an increase in O-GlcNAc signaling in macrophages. O-GlcNAc signaling is down-regulated during macrophage proinflammatory activation. Suppressing O-GlcNAc signaling by O-GlcNAc transferase (OGT) knockout enhances macrophage proinflammatory polarization, promotes adipose tissue inflammation and lipolysis, increases lipid accumulation in peripheral tissues, and exacerbates tissue-specific and whole-body insulin resistance in high-fat-diet-induced obese mice. OGT inhibits macrophage proinflammatory activation by catalyzing ribosomal protein S6 kinase beta-1 (S6K1) O-GlcNAcylation and suppressing S6K1 phosphorylation and mTORC1 signaling. These findings thus identify macrophage O-GlcNAc signaling as a homeostatic mechanism maintaining whole-body metabolism under overnutrition.


Subject(s)
Macrophages/immunology , N-Acetylglucosaminyltransferases/immunology , Obesity/immunology , Ribosomal Protein S6 Kinases, 90-kDa/immunology , Acetylglucosamine/immunology , Adipose Tissue/immunology , Animals , Humans , Macrophage Activation , Macrophages/enzymology , Mice , Mice, Knockout , N-Acetylglucosaminyltransferases/genetics , Obesity/enzymology , Obesity/genetics , Obesity/metabolism , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Signal Transduction
20.
Annu Rev Pharmacol Toxicol ; 59: 65-87, 2019 01 06.
Article in English | MEDLINE | ID: mdl-30625285

ABSTRACT

Type 2 diabetes (T2D) is characterized by persistent hyperglycemia despite hyperinsulinemia, affects more than 400 million people worldwide, and is a major cause of morbidity and mortality. Insulin resistance, of which ectopic lipid accumulation in the liver [nonalcoholic fatty liver disease (NAFLD)] and skeletal muscle is the root cause, plays a major role in the development of T2D. Although lifestyle interventions and weight loss are highly effective at reversing NAFLD and T2D, weight loss is difficult to sustain, and newer approaches aimed at treating the root cause of T2D are urgently needed. In this review, we highlight emerging pharmacological strategies aimed at improving insulin sensitivity and T2D by altering hepatic energy balance or inhibiting key enzymes involved in hepatic lipid synthesis. We also summarize recent research suggesting that liver-targeted mitochondrial uncoupling may be an attractive therapeutic approach to treat NAFLD, nonalcoholic steatohepatitis, and T2D.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Insulin Resistance/physiology , Non-alcoholic Fatty Liver Disease/drug therapy , Animals , Energy Metabolism/drug effects , Humans , Liver/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL