Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Nat Commun ; 15(1): 3682, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38693121

ABSTRACT

In diabetes, macrophages and inflammation are increased in the islets, along with ß-cell dysfunction. Here, we demonstrate that galectin-3 (Gal3), mainly produced and secreted by macrophages, is elevated in islets from both high-fat diet (HFD)-fed and diabetic db/db mice. Gal3 acutely reduces glucose-stimulated insulin secretion (GSIS) in ß-cell lines and primary islets in mice and humans. Importantly, Gal3 binds to calcium voltage-gated channel auxiliary subunit gamma 1 (CACNG1) and inhibits calcium influx via the cytomembrane and subsequent GSIS. ß-Cell CACNG1 deficiency phenocopies Gal3 treatment. Inhibition of Gal3 through either genetic or pharmacologic loss of function improves GSIS and glucose homeostasis in both HFD-fed and db/db mice. All animal findings are applicable to male mice. Here we show a role of Gal3 in pancreatic ß-cell dysfunction, and Gal3 could be a therapeutic target for the treatment of type 2 diabetes.


Subject(s)
Diet, High-Fat , Galectin 3 , Insulin Secretion , Insulin-Secreting Cells , Animals , Humans , Male , Mice , Calcium/metabolism , Calcium Channels/metabolism , Calcium Channels/genetics , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/genetics , Diet, High-Fat/adverse effects , Galectin 3/metabolism , Galectin 3/genetics , Glucose/metabolism , Insulin/metabolism , Insulin Secretion/drug effects , Insulin-Secreting Cells/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout
2.
Nat Commun ; 14(1): 6991, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37914684

ABSTRACT

Follicle-stimulating hormone (FSH) is involved in mammalian reproduction via binding to FSH receptor (FSHR). However, several studies have found that FSH and FSHR play important roles in extragonadal tissue. Here, we identified the expression of FSHR in human and mouse pancreatic islet ß-cells. Blocking FSH signaling by Fshr knock-out led to impaired glucose tolerance owing to decreased insulin secretion, while high FSH levels caused insufficient insulin secretion as well. In vitro, we found that FSH orchestrated glucose-stimulated insulin secretion (GSIS) in a bell curve manner. Mechanistically, FSH primarily activates Gαs via FSHR, promoting the cAMP/protein kinase A (PKA) and calcium pathways to stimulate GSIS, whereas high FSH levels could activate Gαi to inhibit the cAMP/PKA pathway and the amplified effect on GSIS. Our results reveal the role of FSH in regulating pancreatic islet insulin secretion and provide avenues for future clinical investigation and therapeutic strategies for postmenopausal diabetes.


Subject(s)
Follicle Stimulating Hormone , Islets of Langerhans , Mice , Animals , Humans , Follicle Stimulating Hormone/pharmacology , Follicle Stimulating Hormone/metabolism , Insulin Secretion , Glucose/pharmacology , Glucose/metabolism , Receptors, FSH/genetics , Receptors, FSH/metabolism , Islets of Langerhans/metabolism , Signal Transduction , Insulin/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Mammals/metabolism
3.
Diabetes ; 72(11): 1574-1596, 2023 Nov 01.
Article in English | MEDLINE | ID: mdl-37579296

ABSTRACT

Thermogenic adipocytes have been extensively investigated because of their energy-dissipating property and therapeutic potential for obesity and diabetes. Besides serving as fuel sources, accumulating evidence suggests that intermediate metabolites play critical roles in multiple biological processes. However, their role in adipocyte differentiation and thermogenesis remains unexplored. Here, we report that human and mouse obesity is associated with marked downregulation of glutamine synthetase (Glul) expression and activity in thermogenic adipose tissues. Glul is robustly upregulated during brown adipocyte (BAC) differentiation and in brown adipose tissue (BAT) upon cold exposure and Cl316,243 stimulation. Further genetic, pharmacologic, or metabolic manipulations of Glul and glutamine levels reveal that glutamine cells autonomously stimulate BAC differentiation and function and BAT remodeling and improve systemic energy homeostasis in mice. Mechanistically, glutamine promotes transcriptional induction of adipogenic and thermogenic gene programs through histone modification-mediated chromatin remodeling. Among all the glutamine-regulated writer and eraser genes responsible for histone methylation and acetylation, only Prdm9, a histone lysine methyltransferase, is robustly induced during BAC differentiation. Importantly, Prdm9 inactivation by shRNA knockdown or a selective inhibitor attenuates glutamine-triggered adipogenic and thermogenic induction. Furthermore, Prdm9 gene transcription is regulated by glutamine through the recruitment of C/EBPb to its enhancer region. This work reveals glutamine as a novel activator of thermogenic adipocyte differentiation and uncovers an unexpected role of C/EBPb-Prdm9-mediated H3K4me3 and transcriptional reprogramming in adipocyte differentiation and thermogenesis.

4.
Nat Commun ; 14(1): 4257, 2023 07 19.
Article in English | MEDLINE | ID: mdl-37468484

ABSTRACT

Skeletal muscle and thermogenic adipose tissue are both critical for the maintenance of body temperature in mammals. However, whether these two tissues are interconnected to modulate thermogenesis and metabolic homeostasis in response to thermal stress remains inconclusive. Here, we report that human and mouse obesity is associated with elevated Musclin levels in both muscle and circulation. Intriguingly, muscle expression of Musclin is markedly increased or decreased when the male mice are housed in thermoneutral or chronic cool conditions, respectively. Beige fat is then identified as the primary site of Musclin action. Muscle-transgenic or AAV-mediated overexpression of Musclin attenuates beige fat thermogenesis, thereby exacerbating diet-induced obesity and metabolic disorders in male mice. Conversely, Musclin inactivation by muscle-specific ablation or neutralizing antibody treatment promotes beige fat thermogenesis and improves metabolic homeostasis in male mice. Mechanistically, Musclin binds to transferrin receptor 1 (Tfr1) and antagonizes Tfr1-mediated cAMP/PKA-dependent thermogenic induction in beige adipocytes. This work defines the temperature-sensitive myokine Musclin as a negative regulator of adipose thermogenesis that exacerbates the deterioration of metabolic health in obese male mice and thus provides a framework for the therapeutic targeting of this endocrine pathway.


Subject(s)
Adipose Tissue, Beige , Adipose Tissue, White , Animals , Humans , Male , Mice , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Homeostasis , Mammals , Mice, Inbred C57BL , Muscles/metabolism , Obesity/metabolism , Thermogenesis
5.
J Exp Med ; 220(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37284884

ABSTRACT

Obesity and type 2 diabetes (T2D) are the leading causes of the progressive decline in muscle regeneration and fitness in adults. The muscle microenvironment is known to play a key role in controlling muscle stem cell regenerative capacity, yet the underlying mechanism remains elusive. Here, we found that Baf60c expression in skeletal muscle is significantly downregulated in obese and T2D mice and humans. Myofiber-specific ablation of Baf60c in mice impairs muscle regeneration and contraction, accompanied by a robust upregulation of Dkk3, a muscle-enriched secreted protein. Dkk3 inhibits muscle stem cell differentiation and attenuates muscle regeneration in vivo. Conversely, Dkk3 blockade by myofiber-specific Baf60c transgene promotes muscle regeneration and contraction. Baf60c interacts with Six4 to synergistically suppress myocyte Dkk3 expression. While muscle expression and circulation levels of Dkk3 are markedly elevated in obese mice and humans, Dkk3 knockdown improves muscle regeneration in obese mice. This work defines Baf60c in myofiber as a critical regulator of muscle regeneration through Dkk3-mediated paracrine signaling.


Subject(s)
Diabetes Mellitus, Type 2 , Paracrine Communication , Humans , Adult , Mice , Animals , Diabetes Mellitus, Type 2/metabolism , Mice, Obese , Muscle, Skeletal/metabolism , Regeneration
6.
Cell Metab ; 35(4): 711-721.e4, 2023 04 04.
Article in English | MEDLINE | ID: mdl-37019081

ABSTRACT

Metabolism is fundamental to life, but measuring metabolic reaction rates remains challenging. Here, we applied C13 fluxomics to monitor the metabolism of dietary glucose carbon in 12 tissues, 9 brain compartments, and over 1,000 metabolite isotopologues over a 4-day period. The rates of 85 reactions surrounding central carbon metabolism are determined with elementary metabolite unit (EMU) modeling. Lactate oxidation, not glycolysis, occurs at a comparable pace with the tricarboxylic acid cycle (TCA), supporting lactate as the primary fuel. We expand the EMU framework to track and quantify metabolite flows across tissues. Specifically, multi-organ EMU simulation of uridine metabolism shows that tissue-blood exchange, not synthesis, controls nucleotide homeostasis. In contrast, isotopologue fingerprinting and kinetic analyses reveal the brown adipose tissue (BAT) having the highest palmitate synthesis activity but no apparent contribution to circulation, suggesting a tissue-autonomous synthesis-to-burn mechanism. Together, this study demonstrates the utility of dietary fluxomics for kinetic mapping in vivo and provides a rich resource for elucidating inter-organ metabolic cross talk.


Subject(s)
Carbon , Glucose , Animals , Mice , Glucose/metabolism , Carbon/metabolism , Citric Acid Cycle , Lactic Acid/metabolism , Lipids
7.
J Mol Cell Biol ; 15(3)2023 08 03.
Article in English | MEDLINE | ID: mdl-36882217

ABSTRACT

Exercise intervention at the early stage of type 2 diabetes mellitus (T2DM) can aid in the maintenance of blood glucose homeostasis and prevent the development of macrovascular and microvascular complications. However, the exercise-regulated pathways that prevent the development of T2DM remain largely unclear. In this study, two forms of exercise intervention, treadmill training and voluntary wheel running, were conducted for high-fat diet (HFD)-induced obese mice. We observed that both forms of exercise intervention alleviated HFD-induced insulin resistance and glucose intolerance. Skeletal muscle is recognized as the primary site for postprandial glucose uptake and for responsive alteration beyond exercise training. Metabolomic profiling of the plasma and skeletal muscle in Chow, HFD, and HFD-exercise groups revealed robust alterations in metabolic pathways by exercise intervention in both cases. Overlapping analysis identified nine metabolites, including beta-alanine, leucine, valine, and tryptophan, which were reversed by exercise treatment in both the plasma and skeletal muscle. Transcriptomic analysis of gene expression profiles in the skeletal muscle revealed several key pathways involved in the beneficial effects of exercise on metabolic homeostasis. In addition, integrative transcriptomic and metabolomic analyses uncovered strong correlations between the concentrations of bioactive metabolites and the expression levels of genes involved in energy metabolism, insulin sensitivity, and immune response in the skeletal muscle. This work established two models of exercise intervention in obese mice and provided mechanistic insights into the beneficial effects of exercise intervention on systemic energy homeostasis.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Mice , Animals , Humans , Transcriptome , Mice, Obese , Diabetes Mellitus, Type 2/metabolism , Motor Activity , Diet, High-Fat/adverse effects , Metabolome , Muscle, Skeletal/metabolism , Exercise Therapy , Mice, Inbred C57BL
8.
Sci Bull (Beijing) ; 67(7): 733-747, 2022 04 15.
Article in English | MEDLINE | ID: mdl-36546138

ABSTRACT

Diabetes is caused by the interplay between genetics and environmental factors, tightly linked to lifestyle and dietary patterns. In this study, we explored the effectiveness of intermittent protein restriction (IPR) in diabetes control. IPR drastically reduced hyperglycemia in both streptozotocin-treated and leptin receptor-deficient db/db mouse models. IPR improved the number, proliferation, and function of ß cells in pancreatic islets. IPR reduced glucose production in the liver and elevated insulin signaling in the skeletal muscle. IPR elevated serum level of FGF21, and deletion of the Fgf21 gene in the liver abrogated the hypoglycemic effect of IPR without affecting ß cells. IPR caused less lipid accumulation and damage in the liver than that caused by continuous protein restriction in streptozotocin-treated mice. Single-cell RNA sequencing using mouse islets revealed that IPR reversed diabetes-associated ß cell reduction and immune cell accumulation. As IPR is not based on calorie restriction and is highly effective in glycemic control and ß cell protection, it has promising translational potential in the future.


Subject(s)
Diabetes Mellitus, Experimental , Islets of Langerhans , Mice , Animals , Diabetes Mellitus, Experimental/metabolism , Diet, Protein-Restricted , Streptozocin/metabolism , Glucose/metabolism , Homeostasis
9.
Yi Chuan ; 44(10): 840-852, 2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36384722

ABSTRACT

In recent years, the incidence rate of type 2 diabetes (T2D) has risen rapidly and has become a global health crisis. Recent experimental and clinical studies have shown that islet ß-cell dysfunction is an important cause of T2D and its related complications. ß-cells undergo dynamic compensation and decompensation in the course of T2D. In this process, metabolic stress responses, such as ER stress, oxidative stress and inflammation, are key regulators of ß-cell functional alternations. In this review, we summarize the research progress on the ß-cell functional dynamics in the course of T2D, in order to deepen the understanding of the molecular mechanism of T2D, and provide reference for its precise diagnosis and clinical intervention.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Islets of Langerhans , Humans , Islets of Langerhans/metabolism , Insulin-Secreting Cells/metabolism , Inflammation , Oxidative Stress
10.
Yi Chuan ; 44(10): 958-966, 2022 Oct 20.
Article in English | MEDLINE | ID: mdl-36384731

ABSTRACT

Cleavage under target and tagment (CUT&Tag) is a technology that utilizes the fusion protein of Tn5 transposase and protein A/G which can guide Tn5 enzyme to the antibody bound to target protein and cleave the chromatin regions adjacent to target protein. Chromatin libraries are then tagged and sequenced by the high-throughput sequencing to obtain chromatin information at specific sites or protein binding locations. CUT&Tag technology plays an important role in the research of DNA and protein interactions. It can be used to understand the modifications of histone and the bindings of transcription factors. Compared with the traditional chromatin immunoprecipitation-sequencing (ChIP-seq) technology, the CUT&Tag has the strengths of high signal-to-noise ratio, good repeatability, short experimental period, and low cell input. It shows great advantages in early embryonic development, stem cells, cancer, epigenetics and other research fields. In this article, we described the protocol of CUT&Tag for metabolic tissue cells (mouse primary islet cells), to provide an epigenetic method for studying metabolic cells.


Subject(s)
Chromatin , Histones , Mice , Animals , Chromatin Immunoprecipitation/methods , Sequence Analysis, DNA/methods , Histones/metabolism , High-Throughput Nucleotide Sequencing
12.
J Mol Cell Biol ; 14(7)2022 11 30.
Article in English | MEDLINE | ID: mdl-35929791

ABSTRACT

Pancreatic ß-cell dysfunction and insulin resistance are two of the major causes of type 2 diabetes (T2D). Recent clinical and experimental studies have suggested that the functional capacity of ß-cells, particularly in the first phase of insulin secretion, is a primary contributor to the progression of T2D and its associated complications. Pancreatic ß-cells undergo dynamic compensation and decompensation processes during the development of T2D, in which metabolic stresses such as endoplasmic reticulum stress, oxidative stress, and inflammatory signals are key regulators of ß-cell dynamics. Dietary and exercise interventions have been shown to be effective approaches for the treatment of obesity and T2D, especially in the early stages. Whilst the targeted tissues and underlying mechanisms of dietary and exercise interventions remain somewhat vague, accumulating evidence has implicated the improvement of ß-cell functional capacity. In this review, we summarize recent advances in the understanding of the dynamic adaptations of ß-cell function in T2D progression and clarify the effects and mechanisms of dietary and exercise interventions on ß-cell dysfunction in T2D. This review provides molecular insights into the therapeutic effects of dietary and exercise interventions on T2D, and more importantly, it paves the way for future research on the related underlying mechanisms for developing precision prevention and treatment of T2D.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Insulin-Secreting Cells , Humans , Diabetes Mellitus, Type 2/therapy , Diabetes Mellitus, Type 2/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Exercise Therapy/adverse effects
13.
Diabetes ; 71(10): 2136-2152, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35822944

ABSTRACT

Adipose tissue macrophage (ATM) has been shown to play a key role in the pathogenesis of obesity-associated adipose tissue inflammation and metabolic diseases. However, the upstream factors that integrate the environmental signals to control ATM activation and adipose inflammation in obesity remain elusive. Here, we identify BAF60a, a subunit of the switch/sucrose-nonfermentable (SWI/SNF) chromatin remodeling complexes, as the central checkpoint regulator of obesity-induced ATM activation, adipose tissue inflammation, and systemic metabolic impairment. BAF60a expression was robustly downregulated in the adipose tissue stromal vascular fractions in type 2 diabetic mice. Myeloid-specific BAF60a knockout (BaMKO) promotes ATM proinflammatory activation, exacerbating diet-induced obesity, insulin resistance, and metabolic dysfunction. Conversely, myeloid-specific overexpression of BAF60a in mice attenuates macrophage proinflammatory activation. Mechanistically, transcriptome and chromatin landscape analyses demonstrate that BAF60a inactivation triggers the expression of proinflammatory gene program through chromatin remodeling. Moreover, motif analysis of ATAC-Seq and CUT&Tag-Seq data identifies the transcription factor Atf3 that physically interacts with BAF60a to suppress the proinflammatory gene expression, thereby controlling ATM activation and metabolic inflammation in obesity. Consistently, myeloid-specific Atf3 deficiency also promotes the proinflammatory activation of macrophage. This work uncovers BAF60a/Atf3 axis as the key regulator in obesity-associated ATM activation, adipose tissue inflammation, and metabolic diseases.


Subject(s)
Diabetes Mellitus, Experimental , Insulin Resistance , Adipose Tissue/metabolism , Animals , Chromatin/metabolism , Chromosomal Proteins, Non-Histone , Diabetes Mellitus, Experimental/metabolism , Diet , Inflammation/genetics , Inflammation/metabolism , Insulin Resistance/genetics , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/metabolism , Sucrose/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
J Exp Med ; 219(7)2022 07 04.
Article in English | MEDLINE | ID: mdl-35652891

ABSTRACT

Pancreatic ß cell plasticity is the primary determinant of disease progression and remission of type 2 diabetes (T2D). However, the dynamic nature of ß cell adaptation remains elusive. Here, we establish a mouse model exhibiting the compensation-to-decompensation adaptation of ß cell function in response to increasing duration of high-fat diet (HFD) feeding. Comprehensive islet functional and transcriptome analyses reveal a dynamic orchestration of transcriptional networks featuring temporal alteration of chromatin remodeling. Interestingly, prediabetic dietary intervention completely rescues ß cell dysfunction, accompanied by a remarkable reversal of HFD-induced reprogramming of islet chromatin accessibility and transcriptome. Mechanistically, ATAC-based motif analysis identifies CTCF as the top candidate driving dietary intervention-induced preservation of ß cell function. CTCF expression is markedly decreased in ß cells from obese and diabetic mice and humans. Both dietary intervention and AAV-mediated restoration of CTCF expression ameliorate ß cell dysfunction ex vivo and in vivo, through transducing the lipid toxicity and inflammatory signals to transcriptional reprogramming of genes critical for ß cell glucose metabolism and stress response.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Animals , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Diet, High-Fat , Insulin-Secreting Cells/metabolism , Mice , Obesity/genetics , Obesity/metabolism
15.
J Genet Genomics ; 49(4): 316-328, 2022 04.
Article in English | MEDLINE | ID: mdl-35292418

ABSTRACT

Type 2 diabetes (T2D) is caused by insulin resistance and insufficient insulin secretion. Evidence has increasingly indicated that pancreatic ß-cell dysfunction is the primary determinant of T2D disease progression and remission. High plasticity is an important feature of pancreatic ß-cells. During T2D development, pancreatic ß-cells undergo dynamic adaptation. Although ß-cell death/apoptosis in later-stage T2D is the major cause of ß-cell dysfunction, recent studies have revealed that ß-cell dedifferentiation and reprogramming, which play critical roles in ß-cell functional regulation in the early and middle T2D progression stages, are characterized by (i) a loss of mature ß-cell-enriched genes; (ii) dedifferentiation to a progenitor-like state; and (iii) transdifferentiation into other cell types. The roles of transcription factors (TFs) in the establishment and maintenance of ß-cell identity during pancreatic development have been extensively studied. Here, we summarize the roles and underlying mechanisms of TFs in the maintenance of ß-cell identity under physiological and type 2 diabetic conditions. Several feasible approaches for restoring islet functions are also discussed. A better understanding of the transcriptional control of ß-cell identity and plasticity will pave the way for developing more effective strategies, such as ß-cell regeneration therapy, to treat T2D and associated metabolic disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin-Secreting Cells , Cell Dedifferentiation/genetics , Diabetes Mellitus, Type 2/metabolism , Gene Expression Regulation , Humans , Insulin/genetics , Insulin/metabolism , Insulin Secretion/genetics , Insulin-Secreting Cells/metabolism
16.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Article in English | MEDLINE | ID: mdl-35145023

ABSTRACT

Insulin resistance and ß-cell dysfunction are two main molecular bases yet to be further elucidated for type 2 diabetes (T2D). Accumulating evidence indicates that stimulator of interferon genes (STING) plays an important role in regulating insulin sensitivity. However, its function in ß-cells remains unknown. Herein, using global STING knockout (STING-/-) and ß-cell-specific STING knockout (STING-ßKO) mouse models, we revealed a distinct role of STING in the regulation of glucose homeostasis through peripheral tissues and ß-cells. Specially, although STING-/- beneficially alleviated insulin resistance and glucose intolerance induced by high-fat diet, it surprisingly impaired islet glucose-stimulated insulin secretion (GSIS). Importantly, STING is decreased in islets of db/db mice and patients with T2D, suggesting a possible role of STING in ß-cell dysfunction. Indeed, STING-ßKO caused glucose intolerance due to impaired GSIS, indicating that STING is required for normal ß-cell function. Islet transcriptome analysis showed that STING deficiency decreased expression of ß-cell function-related genes, including Glut2, Kcnj11, and Abcc8, contributing to impaired GSIS. Mechanistically, the assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and cleavage under targets and tagmentation (CUT&Tag) analyses suggested that Pax6 was the transcription factor that might be associated with defective GSIS in STING-ßKO mice. Indeed, Pax6 messenger RNA and protein levels were down-regulated and its nuclear localization was lost in STING-ßKO ß-cells. Together, these data revealed a function of STING in the regulation of insulin secretion and established pathophysiological significance of fine-tuned STING within ß-cells and insulin target tissues for maintaining glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 2/metabolism , Glucose Intolerance/chemically induced , Glucose/metabolism , Insulin/metabolism , Membrane Proteins/metabolism , Animals , Diabetes Mellitus, Experimental , Diet, High-Fat/adverse effects , Down-Regulation , Gene Expression Regulation , Homeostasis , Humans , Insulin/blood , Insulin Resistance , Insulin-Secreting Cells , Membrane Proteins/genetics , Mice , Mice, Knockout
17.
Cell Death Differ ; 28(12): 3214-3234, 2021 12.
Article in English | MEDLINE | ID: mdl-34045668

ABSTRACT

Dysregulation of glucose homeostasis contributes to insulin resistance and type 2 diabetes. Whilst exercise stimulated activation of AMP-activated protein kinase (AMPK), an important energy sensor, has been highlighted for its potential to promote insulin-stimulated glucose uptake, the underlying mechanisms for this remain largely unknown. Here we found that AMPK positively regulates the activation of Rab5, a small GTPase which is involved in regulating Glut4 translocation, in both myoblasts and skeletal muscles. We further verified that TBC1D17, identified as a potential interacting partner of Rab5 in our recent study, is a novel GTPase activating protein (GAP) of Rab5. TBC1D17-Rab5 axis regulates transport of Glut1, Glut4, and transferrin receptor. TBC1D17 interacts with Rab5 or AMPK via its TBC domain or N-terminal 1-306 region (N-Ter), respectively. Moreover, AMPK phosphorylates the Ser 168 residue of TBC1D17 which matches the predicted AMPK consensus motif. N-Ter of TBC1D17 acts as an inhibitory region by directly interacting with the TBC domain. Ser168 phosphorylation promotes intra-molecular interaction and therefore enhances the auto-inhibition of TBC1D17. Our findings reveal that TBC1D17 acts as a molecular bridge that links AMPK and Rab5 and delineate a previously unappreciated mechanism by which the activation of TBC/RabGAP is regulated.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Diabetes Mellitus, Type 2/genetics , GTPase-Activating Proteins/metabolism , Glucose/metabolism , Insulin Resistance/genetics , rab5 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Animals , Diabetes Mellitus, Type 2/pathology , Humans , Male , Mice , Phosphorylation , Transfection
18.
Metabol Open ; 10: 100088, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33855290
19.
Liver Int ; 41(4): 777-787, 2021 04.
Article in English | MEDLINE | ID: mdl-33555112

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD), whose pathogenesis remains unelucidated, has become an increasingly prevalent disease globally requiring novel treatment strategies. This study aims to explore the role of leukocyte cell-derived chemotaxin 2 (LECT2), one of the known hepatokines, in the development of NAFLD. METHODS: The serum LECT2 level was evaluated in patients with NAFLD and male C57BL/6 mice fed a high-fat diet (HFD) for 8 weeks. Tail intravenous injection of adeno-associated virus that contained Lect2 short hairpin RNA or Lect2 overexpression plasmid was administered to mice to inhibit or increase hepatic Lect2 expression. Hepatic steatosis was evaluated by histological staining with haematoxylin and eosin and Oil Red O, and also by quantitative hepatic triglyceride measurements. RNA-seq was performed to discover the specific targets of LECT2 on NAFLD. RESULTS: Serum and hepatic LECT2 levels were elevated in NAFLD patients and HFD-fed mice. Inhibition of hepatic Lect2 expression alleviated HFD-induced hepatic steatosis and inflammation, whereas hepatic overexpression of Lect2 aggravated HFD-induced hepatic steatosis and inflammation. RNA-seq and bioinformatical analysis suggested that the signal transducers and activators of transcription-1 (STAT-1) pathway might play an indispensable role in the interaction between LECT2 and NAFLD. A STAT-1 inhibitor could reverse the accumulation of hepatic lipids caused by Lect2 overexpression. CONCLUSION: LECT2 expression is significantly elevated in NAFLD. LECT2 induces the occurrence and development of NAFLD through the STAT-1 pathway. LECT2 may be a potential therapeutic target for NAFLD.


Subject(s)
Intercellular Signaling Peptides and Proteins , Non-alcoholic Fatty Liver Disease , Animals , Chemotactic Factors , Diet, High-Fat , Humans , Intercellular Signaling Peptides and Proteins/genetics , Leukocytes , Liver , Male , Mice , Mice, Inbred C57BL , Transducers
20.
Clin Nutr ; 40(4): 2025-2034, 2021 04.
Article in English | MEDLINE | ID: mdl-33008652

ABSTRACT

BACKGROUND & AIMS: Our previous study found that platelet counts were positively associated with body fat percentage in human. In the present study, we conducted a reverse translational study to explore the role of platelets in modulating pre-adipocyte proliferation in mice. METHODS: Mouse pre-adipocyte cell line (3T3-L1) and human pre-adipocytes harvested from female subcutaneous fat were used. Pre-adipocytes were co-cultured with platelets or platelet releasate, which were isolated from mice or humans. The cell viability and proliferative ability of the pre-adipocytes were examined by MTT and flow cytometry assays. Western blotting analysis was used to determine the phosphorylation levels of proteins in the mTOR pathway. RESULTS: The number of platelets in the adipose tissues from obese mice was significantly higher than that from lean mice. Platelets and collagen-activated platelet releasate stimulated the proliferation of human pre-adipocytes and 3T3-L1 cells in vitro. Besides, platelets from obese mice were more potent in stimulating pre-adipocyte proliferation than those from lean control mice. Mechanistically, platelets enhanced pre-adipocyte proliferation through the acceleration of cell cycle progression from G0/G1 to S phase cell cycle progression. At the molecular level, platelets promoted pre-adipocyte proliferation through mTOR pathway-mediated upregulation of cyclin D1 expression. CONCLUSION: In conclusion, platelets and platelet releasate play an important role in the proliferation of pre-adipocytes. Our study may provide new clues and the molecular mechanism of the causal pathways between platelets and body fat to explain the finding we observed in population study.


Subject(s)
Adipocytes/physiology , Adipose Tissue/physiopathology , Blood Platelets/physiology , 3T3-L1 Cells , Adipose Tissue/pathology , Animals , Apoptosis , Blood Platelets/pathology , Cell Communication , Cell Proliferation , Cell Survival , Cells, Cultured , Cyclin D1/physiology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/pathology , Obesity/physiopathology , Platelet Count , Specific Pathogen-Free Organisms , Subcutaneous Fat , TOR Serine-Threonine Kinases/physiology , Translational Research, Biomedical
SELECTION OF CITATIONS
SEARCH DETAIL
...