Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Cell ; 184(13): 3502-3518.e33, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34048700

ABSTRACT

Thermogenic adipocytes possess a therapeutically appealing, energy-expending capacity, which is canonically cold-induced by ligand-dependent activation of ß-adrenergic G protein-coupled receptors (GPCRs). Here, we uncover an alternate paradigm of GPCR-mediated adipose thermogenesis through the constitutively active receptor, GPR3. We show that the N terminus of GPR3 confers intrinsic signaling activity, resulting in continuous Gs-coupling and cAMP production without an exogenous ligand. Thus, transcriptional induction of Gpr3 represents the regulatory parallel to ligand-binding of conventional GPCRs. Consequently, increasing Gpr3 expression in thermogenic adipocytes is alone sufficient to drive energy expenditure and counteract metabolic disease in mice. Gpr3 transcription is cold-stimulated by a lipolytic signal, and dietary fat potentiates GPR3-dependent thermogenesis to amplify the response to caloric excess. Moreover, we find GPR3 to be an essential, adrenergic-independent regulator of human brown adipocytes. Taken together, our findings reveal a noncanonical mechanism of GPCR control and thermogenic activation through the lipolysis-induced expression of constitutively active GPR3.


Subject(s)
Adipose Tissue, Brown/metabolism , Constitutive Androstane Receptor/metabolism , Lipolysis , Receptors, G-Protein-Coupled/metabolism , Thermogenesis , Adipocytes/metabolism , Animals , COS Cells , Cells, Cultured , Chlorocebus aethiops , Cold Temperature , Dietary Fats/pharmacology , Humans , Mice, Inbred C57BL , Phenotype , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Sympathetic Nervous System/metabolism , Transcription, Genetic
2.
J Physiol ; 600(4): 885-902, 2022 02.
Article in English | MEDLINE | ID: mdl-34387373

ABSTRACT

KEY POINTS: Afadin is a ubiquitously expressed scaffold protein with a recently discovered role in insulin signalling and glucose metabolism. Insulin-stimulated phosphorylation of Afadin at S1795 occurs in insulin-responsive tissues such as adipose tissue, muscle, liver, pancreas and heart. Afadin abundance and AfadinS1795 phosphorylation are dynamically regulated in metabolic tissues during diet-induced obesity progression. Genetic silencing of AfadinS1795 phosphorylation improves glucose homeostasis in the early stages of diet-induced metabolic dysregulation. AfadinS1795 phosphorylation contributes to the early development of obesity-related complications in mice. ABSTRACT: Obesity is associated with systemic insulin resistance and numerous metabolic disorders. Yet, the mechanisms underlying impaired insulin action during obesity remain to be fully elucidated. Afadin is a multifunctional scaffold protein with the ability to modulate insulin action through its phosphorylation at S1795 in adipocytes. In the present study, we report that insulin-stimulated AfadinS1795 phosphorylation is not restricted to adipose tissues, but is a common signalling event in insulin-responsive tissues including muscle, liver, pancreas and heart. Furthermore, a dynamic regulation of Afadin abundance occurred during diet-induced obesity progression, while its phosphorylation was progressively attenuated. To investigate the role of AfadinS1795 phosphorylation in the regulation of whole-body metabolic homeostasis, we generated a phospho-defective mouse model (Afadin SA) in which the Afadin phosphorylation site was silenced (S1795A) at the whole-body level using CRISPR-Cas9-mediated gene editing. Metabolic characterization of these mice under basal physiological conditions or during a high-fat diet (HFD) challenge revealed that preventing AfadinS1795 phosphorylation improved insulin sensitivity and glucose tolerance and increased liver glycogen storage in the early stage of diet-induced metabolic dysregulation, without affecting body weight. Together, our findings reveal that AfadinS1795 phosphorylation in metabolic tissues is critical during obesity progression and contributes to promote systemic insulin resistance and glucose intolerance in the early phase of diet-induced obesity.


Subject(s)
Insulin Resistance , Animals , Diet, High-Fat , Glucose/metabolism , Homeostasis , Insulin/metabolism , Insulin Resistance/physiology , Mice , Mice, Inbred C57BL , Mice, Obese , Microfilament Proteins , Phosphorylation
3.
Chemistry ; 27(9): 3166-3176, 2021 Feb 10.
Article in English | MEDLINE | ID: mdl-33169429

ABSTRACT

A glucose responsive insulin (GRI) that responds to changes in blood glucose concentrations has remained an elusive goal. Here we describe the development of glucose cleavable linkers based on hydrazone and thiazolidine structures. We developed linkers with low levels of spontaneous hydrolysis but increased level of hydrolysis with rising concentrations of glucose, which demonstrated their glucose responsiveness in vitro. Lipidated hydrazones and thiazolidines were conjugated to the LysB29 side-chain of HI by pH-controlled acylations providing GRIs with glucose responsiveness confirmed in vitro for thiazolidines. Clamp studies showed increased glucose infusion at hyperglycemic conditions for one GRI indicative of a true glucose response. The glucose responsive cleavable linker in these GRIs allow changes in glucose levels to drive the release of active insulin from a circulating depot. We have demonstrated an unprecedented, chemically responsive linker concept for biopharmaceuticals.


Subject(s)
Aldehydes/chemistry , Blood Glucose/metabolism , Insulin/chemistry , Insulin/metabolism , Acylation , Animals , Blood Glucose/drug effects , CHO Cells , Cricetulus , Humans , Hydrazones/chemistry , Insulin/pharmacology , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Thiazolidines/chemistry
4.
EMBO Rep ; 20(8): e48216, 2019 08.
Article in English | MEDLINE | ID: mdl-31264358

ABSTRACT

Insulin orchestrates metabolic homeostasis through a complex signaling network for which the precise mechanisms controlling its fine-tuning are not completely understood. Here, we report that Afadin, a scaffold protein, is phosphorylated on S1795 (S1718 in humans) in response to insulin in adipocytes, and this phosphorylation is impaired with obesity and insulin resistance. In turn, loss of Afadin enhances the response to insulin in adipose tissues via upregulation of the insulin receptor protein levels. This happens in a cell-autonomous and phosphorylation-dependent manner. Insulin-stimulated Afadin-S1795 phosphorylation modulates Afadin binding with interaction partners in adipocytes, among which HDAC6 preferentially interacts with phosphorylated Afadin and acts as a key intermediate to suppress insulin receptor protein levels. Adipose tissue-specific Afadin depletion protects against insulin resistance and improves glucose homeostasis in diet-induced obese mice, independently of adiposity. Altogether, we uncover a novel insulin-induced cellular feedback mechanism governed by the interaction of Afadin with HDAC6 to negatively control insulin action in adipocytes, which may offer new strategies to alleviate insulin resistance.


Subject(s)
Adipose Tissue/metabolism , Antigens, CD/genetics , Histone Deacetylase 6/genetics , Insulin/genetics , Microfilament Proteins/genetics , Obesity/genetics , Protein Processing, Post-Translational , Receptor, Insulin/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Antigens, CD/metabolism , Diet, High-Fat/adverse effects , Glucose/metabolism , Histone Deacetylase 6/metabolism , Homeostasis/genetics , Humans , Insulin/metabolism , Insulin/pharmacology , Insulin Resistance , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microfilament Proteins/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Phosphorylation , Primary Cell Culture , Receptor, Insulin/metabolism
5.
Int J Mol Sci ; 21(21)2020 Oct 29.
Article in English | MEDLINE | ID: mdl-33137873

ABSTRACT

Selective inhibition of histone deacetylase 3 (HDAC3) prevents glucolipotoxicity-induced ß-cell dysfunction and apoptosis by alleviation of proapoptotic endoplasmic reticulum (ER) stress-signaling, but the precise molecular mechanisms of alleviation are unexplored. By unbiased microarray analysis of the ß-cell gene expression profile of insulin-producing cells exposed to glucolipotoxicity in the presence or absence of a selective HDAC3 inhibitor, we identified Enhancer of zeste homolog 2 (EZH2) as the sole target candidate. ß-Cells were protected against glucolipotoxicity-induced ER stress and apoptosis by EZH2 attenuation. Small molecule inhibitors of EZH2 histone methyltransferase activity rescued human islets from glucolipotoxicity-induced apoptosis. Moreover, EZH2 knockdown cells were protected against glucolipotoxicity-induced downregulation of the protective non-canonical Nuclear factor of kappa light polypeptide gene enhancer in B-cells (NFκB) pathway. We conclude that EZH2 deficiency protects from glucolipotoxicity-induced ER stress, apoptosis and downregulation of the non-canonical NFκB pathway, but not from insulin secretory dysfunction. The mechanism likely involves transcriptional regulation via EZH2 functioning as a methyltransferase and/or as a methylation-dependent transcription factor.


Subject(s)
Apoptosis , Enhancer of Zeste Homolog 2 Protein/metabolism , Glucose/adverse effects , Insulin Secretion/drug effects , Insulin-Secreting Cells/pathology , Lipids/adverse effects , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/genetics , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Signal Transduction , Sweetening Agents/adverse effects
6.
Mol Carcinog ; 56(10): 2223-2233, 2017 10.
Article in English | MEDLINE | ID: mdl-28544328

ABSTRACT

The pathogenesis of colorectal neoplasia (CRN) has been associated with altered non-neuronal acetylcholine (ACh) metabolism. The aim of this study was to characterize expression, function, and cellular location of ACh-related proteins in biopsies obtained from endoscopic normal-appearing sigmoid colon in patients with and without CRN. Messenger-RNA (mRNA) levels of 17 ACh-related proteins were quantified by rt-qPCR. Functional responses to ACh, measured as electrogenic transepithelial short circuit current (SCC), were recorded using the Ussing chamber technique. Finally, cellular localization of choline transporter-like proteins (CTLs) and butyryl-cholinesterase enzyme (BChE) was determined by immunohistochemistry. mRNA expression of CTL1 and CTL4 was increased in patients with CRN (P = 0.002 and P = 0.04, respectively). In functional experiments, baseline SCC was increased in CRN patients. ACh induced rapid biphasic changes in SCC. An initial decreasing phase was observed in the minority of CRN patients versus the majority of controls (25% vs 69%, respectively, P = 0.031). For the second increasing phase of SCC, data indicated ACh-activation of two receptors. For both parts of the biphasic response, the half maximal effective concentration and maximal responses showed no difference between patient groups. Immunohistochemistry demonstrated CTL1, 3 and 4 and BChE to be localized to colonic crypt cells. We conclude that CRN is associated with increased expression of CTL1 and CTL4, augmented basal prostaglandin-dependent secretion, and altered functional channel response to ACh in human endoscopic normal-appearing colonic mucosa. The immunohistochemical findings support CTL1, CTL3, CTL4, and BChE to be involved in non-neuronal mucosal ACh metabolism.


Subject(s)
Acetylcholine/metabolism , Colorectal Neoplasms/genetics , Gene Expression Profiling/methods , Gene Regulatory Networks , Intestinal Mucosa/metabolism , Aged , Antigens, CD/genetics , Antigens, CD/metabolism , Butyrylcholinesterase/genetics , Butyrylcholinesterase/metabolism , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Middle Aged , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Up-Regulation
7.
Proc Natl Acad Sci U S A ; 111(3): 1055-9, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24395784

ABSTRACT

Type 1 diabetes is due to destruction of pancreatic ß-cells. Lysine deacetylase inhibitors (KDACi) protect ß-cells from inflammatory destruction in vitro and are promising immunomodulators. Here we demonstrate that the clinically well-tolerated KDACi vorinostat and givinostat revert diabetes in the nonobese diabetic (NOD) mouse model of type 1 diabetes and counteract inflammatory target cell damage by a mechanism of action consistent with transcription factor--rather than global chromatin--hyperacetylation. Weaning NOD mice received low doses of vorinostat and givinostat in their drinking water until 100-120 d of age. Diabetes incidence was reduced by 38% and 45%, respectively, there was a 15% increase in the percentage of islets without infiltration, and pancreatic insulin content increased by 200%. Vorinostat treatment increased the frequency of functional regulatory T-cell subsets and their transcription factors Gata3 and FoxP3 in parallel to a decrease in inflammatory dendritic cell subsets and their cytokines IL-6, IL-12, and TNF-α. KDACi also inhibited LPS-induced Cox-2 expression in peritoneal macrophages from C57BL/6 and NOD mice. In insulin-producing ß-cells, givinostat did not upregulate expression of the anti-inflammatory genes Socs1-3 or sirtuin-1 but reduced levels of IL-1ß + IFN-γ-induced proinflammatory Il1a, Il1b, Tnfα, Fas, Cxcl2, and reduced cytokine-induced ERK phosphorylation. Further, NF-κB genomic iNos promoter binding was reduced by 50%, and NF-κB-dependent mRNA expression was blocked. These effects were associated with NF-κB subunit p65 hyperacetylation. Taken together, these data provide a rationale for clinical trials of safety and efficacy of KDACi in patients with autoimmune disease such as type 1 diabetes.


Subject(s)
Chromatin/metabolism , Diabetes Mellitus, Type 1/metabolism , Histone Deacetylase Inhibitors/pharmacology , Insulin-Secreting Cells/cytology , Animals , Cell Line , Cytokines/metabolism , Disease Models, Animal , Epigenesis, Genetic , Female , GATA3 Transcription Factor/metabolism , Histone Deacetylases/metabolism , Humans , Hydroxamic Acids/pharmacology , Inflammation , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Phosphorylation , Promoter Regions, Genetic , Protein Binding , Protein Processing, Post-Translational , Rats , Time Factors , Vorinostat
8.
Int J Mol Sci ; 17(6)2016 Jun 07.
Article in English | MEDLINE | ID: mdl-27338345

ABSTRACT

As microRNAs (miRs) are gaining increasing attention as key regulators of cellular processes, expressional quantification is widely applied. However, in the processing of relatively quantified data, the importance of testing the stability of several reference mRNAs and/or miRs and choosing among these for normalization is often overlooked, potentially leading to biased results. Here, we have optimized the purification of miR-enriched total RNA from pancreatic insulin-producing INS-1 cells. Additionally, we optimized and analyzed miR expression by a qPCR-based microarray and by specific qPCR and tested the stability of candidate reference mRNAs and miRs. Hence, this study gives a widely applicable example on how to easily and systematically test and decide how to normalize miR quantification. We suggest that caution in the interpretation of miR quantification studies that do not comprise stability analysis should be exerted.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Insulin-Secreting Cells/metabolism , MicroRNAs/genetics , Transcriptome , Animals , RNA Stability , Rats , Real-Time Polymerase Chain Reaction , Reproducibility of Results
9.
J Am Chem Soc ; 137(24): 7929-34, 2015 Jun 24.
Article in English | MEDLINE | ID: mdl-26042473

ABSTRACT

Phenotypic cell-based screening is a powerful approach to small-molecule discovery, but a major challenge of this strategy lies in determining the intracellular target and mechanism of action (MoA) for validated hits. Here, we show that the small-molecule BRD0476, a novel suppressor of pancreatic ß-cell apoptosis, inhibits interferon-gamma (IFN-γ)-induced Janus kinase 2 (JAK2) and signal transducer and activation of transcription 1 (STAT1) signaling to promote ß-cell survival. However, unlike common JAK-STAT pathway inhibitors, BRD0476 inhibits JAK-STAT signaling without suppressing the kinase activity of any JAK. Rather, we identified the deubiquitinase ubiquitin-specific peptidase 9X (USP9X) as an intracellular target, using a quantitative proteomic analysis in rat ß cells. RNAi-mediated and CRISPR/Cas9 knockdown mimicked the effects of BRD0476, and reverse chemical genetics using a known inhibitor of USP9X blocked JAK-STAT signaling without suppressing JAK activity. Site-directed mutagenesis of a putative ubiquitination site on JAK2 mitigated BRD0476 activity, suggesting a competition between phosphorylation and ubiquitination to explain small-molecule MoA. These results demonstrate that phenotypic screening, followed by comprehensive MoA efforts, can provide novel mechanistic insights into ostensibly well-understood cell signaling pathways. Furthermore, these results uncover USP9X as a potential target for regulating JAK2 activity in cellular inflammation.


Subject(s)
Insulin-Secreting Cells/drug effects , Interferon-gamma/immunology , Janus Kinase 2/immunology , Protective Agents/chemistry , Protective Agents/pharmacology , STAT1 Transcription Factor/immunology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Humans , Insulin-Secreting Cells/cytology , Insulin-Secreting Cells/immunology , Phosphorylation/drug effects , Rats , Signal Transduction/drug effects , Ubiquitin Thiolesterase/immunology , Ubiquitination/drug effects
10.
J Neuroinflammation ; 11: 27, 2014 Feb 03.
Article in English | MEDLINE | ID: mdl-24490798

ABSTRACT

BACKGROUND: Interleukin 1 (IL-1) is implicated in neuroinflammation, an essential component of neurodegeneration. We evaluated the potential anti-inflammatory effect of a novel peptide antagonist of IL-1 signaling, Ilantide. METHODS: We investigated the binding of Ilantide to IL-1 receptor type I (IL-1RI) using surface plasmon resonance, the inhibition of Il-1ß-induced activation of nuclear factor κB (NF-κB) in HEK-Blue cells that contained an IL-1ß-sensitive reporter, the secretion of TNF-α in macrophages, protection against IL-1-induced apoptosis in neonatal pancreatic islets, and the penetration of Ilantide through the blood-brain barrier using competitive enzyme-linked immunosorbent assay (ELISA). We studied the effects of the peptide on social behavior and memory in rat models of lipopolysaccharide (LPS)- and amyloid-induced neuroinflammation, respectively, and its effect in a rat model of experimental autoimmune enchephalomyelitis. RESULTS: Ilantide bound IL-1RI, inhibited the IL-1ß-induced activation of NF-κB, and inhibited the secretion of TNF-α in vitro. Ilantide protected pancreatic islets from apoptosis in vitro and reduced inflammation in an animal model of arthritis. The peptide penetrated the blood-brain barrier. It reduced the deficits in social activity and memory in LPS- and amyloid-treated animals and delayed the development of experimental autoimmune enchephalomyelitis. CONCLUSIONS: These findings indicate that Ilantide is a novel and potent IL-1RI antagonist that is able to reduce inflammatory damage in the central nervous system and pancreatic islets.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Oligopeptides/pharmacology , Oligopeptides/therapeutic use , Animals , Animals, Newborn , Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Arthritis/chemically induced , Cells, Cultured , Cerebellum/cytology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Humans , Interleukin 1 Receptor Antagonist Protein/chemistry , Interleukin 1 Receptor Antagonist Protein/pharmacology , Lipopolysaccharides , Male , Neurons/drug effects , Neurons/metabolism , Rats , Rats, Wistar , Social Behavior , Transfection , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
11.
Arch Biochem Biophys ; 528(2): 171-84, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-23063755

ABSTRACT

Disease is conventionally viewed as the chaotic inappropriate outcome of deranged tissue function resulting from aberrancies in cellular processes. Yet the patho-biology of cellular dysfunction and death encompasses a coordinated network no less sophisticated and regulated than maintenance of homeostatic balance. Cellular demise is far from passive subordination to stress but requires controlled coordination of energy-requiring activities including gene transcription and protein translation that determine the graded transition between defensive mechanisms, cell cycle regulation, dedifferentiation and ultimately to the activation of death programmes. In fact, most stressors stimulate both homeostasis and regeneration on one hand and impairment and destruction on the other, depending on the ambient circumstances. Here we illustrate this bimodal ambiguity in cell response by reviewing recent progress in our understanding of how the pancreatic ß cell copes with inflammatory stress by changing gene transcription and protein translation by the differential and interconnected action of reactive oxygen and nitric oxide species, microRNAs and posttranslational protein modifications.


Subject(s)
Cytokines/genetics , Cytokines/physiology , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/physiology , Animals , Apoptosis/genetics , Apoptosis/physiology , Histone Deacetylases/metabolism , Humans , Inflammation Mediators/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Models, Biological , Protein Biosynthesis , RNA Processing, Post-Transcriptional , Reactive Oxygen Species/metabolism , Signal Transduction , Transcription, Genetic
12.
BMC Gastroenterol ; 12: 78, 2012 Jun 26.
Article in English | MEDLINE | ID: mdl-22734885

ABSTRACT

BACKGROUND: The pathogenesis of colorectal neoplasia is still unresolved but has been associated with alterations in epithelial clearance of xenobiotics and metabolic waste products. The aim of this study was to functionally characterize the transport of cyclic nucleotides in colonic biopsies from patients with and without colorectal neoplasia. METHODS: Cyclic nucleotides were used as model substrates shared by some OATP- and ABC-transporters, which in part are responsible for clearance of metabolites and xenobiotics from the colonic epithelium. On colonic biopsies from patients with and without colorectal neoplasia, molecular transport was electrophysiologically registered in Ussing-chamber set-ups, mRNA level of selected transporters was quantified by rt-PCR, and subcellular location of transporters was determined by immunohistochemistry. RESULTS: Of four cyclic nucleotides, dibuturyl-cAMP induced the largest short circuit current in both patient groups. The induced short circuit current was significantly lower in neoplasia-patients (p = 0.024). The observed altered transport of dibuturyl-cAMP in neoplasia-patients could not be directly translated to an observed increased mRNA expression of OATP4A1 and OATP2B1 in neoplasia patients. All other examined transporters were expressed to similar extents in both patient groups. CONCLUSIONS: OATP1C1, OATP4A1, OATP4C1 seem to be involved in the excretory system of human colon. ABCC4 is likely to be involved from an endoplasmic-Golgi complex and basolateral location in goblet cells. ABCC5 might be directly involved in the turnover of intracellular cAMP at the basolateral membrane of columnar epithelial cells, while OATP2B1 is indirectly related to the excretory system. Colorectal neoplasia is associated with lower transport or sensitivity to cyclic nucleotides and increased expression of OATP2B1 and OATP4A1 transporters, known to transport PGE(2).


Subject(s)
Colon/metabolism , Colorectal Neoplasms/metabolism , Cyclic AMP/metabolism , Intestinal Mucosa/metabolism , Organic Anion Transporters/metabolism , Aged , Aged, 80 and over , Basement Membrane/metabolism , Colorectal Neoplasms/pathology , Dinoprostone/metabolism , Endoplasmic Reticulum/metabolism , Female , Goblet Cells/metabolism , Golgi Apparatus/metabolism , Humans , Male , Middle Aged , Multidrug Resistance-Associated Proteins/metabolism
13.
Mol Med ; 17(5-6): 378-90, 2011.
Article in English | MEDLINE | ID: mdl-21274504

ABSTRACT

Both common forms of diabetes have an inflammatory pathogenesis in which immune and metabolic factors converge on interleukin-1ß as a key mediator of insulin resistance and ß-cell failure. In addition to improving insulin resistance and preventing ß-cell inflammatory damage, there is evidence of genetic association between diabetes and histone deacetylases (HDACs); and HDAC inhibitors (HDACi) promote ß-cell development, proliferation, differentiation and function and positively affect late diabetic microvascular complications. Here we review this evidence and propose that there is a strong rationale for preclinical studies and clinical trials with the aim of testing the utility of HDACi as a novel therapy for diabetes.


Subject(s)
Diabetes Mellitus/enzymology , Diabetes Mellitus/metabolism , Histone Deacetylases/metabolism , Animals , Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus/drug therapy , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Humans , Interleukin-1beta/metabolism
14.
Sci Rep ; 11(1): 9794, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33963248

ABSTRACT

The profound energy-expending nature of brown adipose tissue (BAT) thermogenesis makes it an attractive target tissue to combat obesity-associated metabolic disorders. While cold exposure is the strongest inducer of BAT activity, the temporal mechanisms tuning BAT adaptation during this activation process are incompletely understood. Here we show that the scaffold protein Afadin is dynamically regulated by cold in BAT, and participates in cold acclimation. Cold exposure acutely increases Afadin protein levels and its phosphorylation in BAT. Knockdown of Afadin in brown pre-adipocytes does not alter adipogenesis but restricts ß3-adrenegic induction of thermogenic genes expression and HSL phosphorylation in mature brown adipocytes. Consistent with a defect in thermogenesis, an impaired cold tolerance was observed in fat-specific Afadin knockout mice. However, while Afadin depletion led to reduced Ucp1 mRNA induction by cold, stimulation of Ucp1 protein was conserved. Transcriptomic analysis revealed that fat-specific ablation of Afadin led to decreased functional enrichment of gene sets controlling essential metabolic functions at thermoneutrality in BAT, whereas it led to an altered reprogramming in response to cold, with enhanced enrichment of different pathways related to metabolism and remodeling. Collectively, we demonstrate a role for Afadin in supporting the adrenergic response in brown adipocytes and BAT function.


Subject(s)
Adipocytes, Brown/metabolism , Adipose Tissue, Brown/metabolism , Cold Temperature , Gene Expression Regulation , Kinesins/biosynthesis , Myosins/biosynthesis , Thermogenesis , Animals , Kinesins/genetics , Mice , Mice, Knockout , Myosins/genetics
15.
Mol Metab ; 44: 101137, 2021 02.
Article in English | MEDLINE | ID: mdl-33285300

ABSTRACT

OBJECTIVE: Increasing adaptive thermogenesis by stimulating browning in white adipose tissue is a promising method of improving metabolic health. However, the molecular mechanisms underlying this transition remain elusive. Our study examined the molecular determinants driving the differentiation of precursor cells into thermogenic adipocytes. METHODS: In this study, we conducted temporal high-resolution proteomic analysis of subcutaneous white adipose tissue (scWAT) after cold exposure in mice. This was followed by loss- and gain-of-function experiments using siRNA-mediated knockdown and CRISPRa-mediated induction of gene expression, respectively, to evaluate the function of the transcriptional regulator Y box-binding protein 1 (YBX1) during adipogenesis of brown pre-adipocytes and mesenchymal stem cells. Transcriptomic analysis of mesenchymal stem cells following induction of endogenous Ybx1 expression was conducted to elucidate transcriptomic events controlled by YBX1 during adipogenesis. RESULTS: Our proteomics analysis uncovered 509 proteins differentially regulated by cold in a time-dependent manner. Overall, 44 transcriptional regulators were acutely upregulated following cold exposure, among which included the cold-shock domain containing protein YBX1, peaking after 24 h. Cold-induced upregulation of YBX1 also occurred in brown adipose tissue, but not in visceral white adipose tissue, suggesting a role of YBX1 in thermogenesis. This role was confirmed by Ybx1 knockdown in brown and brite preadipocytes, which significantly impaired their thermogenic potential. Conversely, inducing Ybx1 expression in mesenchymal stem cells during adipogenesis promoted browning concurrent with an increased expression of thermogenic markers and enhanced mitochondrial respiration. At a molecular level, our transcriptomic analysis showed that YBX1 regulates a subset of genes, including the histone H3K9 demethylase Jmjd1c, to promote thermogenic adipocyte differentiation. CONCLUSION: Our study mapped the dynamic proteomic changes of murine scWAT during browning and identified YBX1 as a novel factor coordinating the genomic mechanisms by which preadipocytes commit to brite/beige lineage.


Subject(s)
Adipose Tissue, White/metabolism , Thermogenesis/genetics , Thermogenesis/physiology , Transcription Factors/genetics , Transcription Factors/metabolism , Adipocytes, Brown/metabolism , Adipogenesis , Adipose Tissue, Brown/metabolism , Animals , Cell Differentiation , Cell Line , Cell Proliferation , Gene Expression Regulation , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Male , Mesenchymal Stem Cells , Mice , Mice, Inbred C57BL , Obesity/metabolism , Proteomics , Subcutaneous Fat/metabolism , Transcriptome , Up-Regulation
16.
Sci Rep ; 11(1): 12253, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112814

ABSTRACT

Loss-of-function (LoF) mutations in KCNQ1, encoding the voltage-gated K+ channel Kv7.1, lead to long QT syndrome 1 (LQT1). LQT1 patients also present with post-prandial hyperinsulinemia and hypoglycaemia. In contrast, KCNQ1 polymorphisms are associated with diabetes, and LQTS patients have a higher prevalence of diabetes. We developed a mouse model with a LoF Kcnq1 mutation using CRISPR-Cas9 and hypothesized that this mouse model would display QT prolongation, increased glucose-stimulated insulin secretion and allow for interrogation of Kv7.1 function in islets. Mice were characterized by electrocardiography and oral glucose tolerance tests. Ex vivo, islet glucose-induced insulin release was measured, and beta-cell area quantified by immunohistochemistry. Homozygous mice had QT prolongation. Ex vivo, glucose-stimulated insulin release was increased in islets from homozygous mice at 12-14 weeks, while beta-cell area was reduced. Non-fasting blood glucose levels were decreased at this age. In follow-up studies 8-10 weeks later, beta-cell area was similar in all groups, while glucose-stimulated insulin secretion was now reduced in islets from hetero- and homozygous mice. Non-fasting blood glucose levels had normalized. These data suggest that Kv7.1 dysfunction is involved in a transition from hyper- to hyposecretion of insulin, potentially explaining the association with both hypoglycemia and hyperglycemia in LQT1 patients.


Subject(s)
Insulin Secretion , Insulin/biosynthesis , Islets of Langerhans/metabolism , KCNQ1 Potassium Channel/genetics , Long QT Syndrome/metabolism , Long QT Syndrome/physiopathology , Loss of Function Mutation , Alleles , Amino Acid Substitution , Animals , Disease Models, Animal , Disease Susceptibility , Glucose/metabolism , Long QT Syndrome/etiology , Mice
17.
Adipocyte ; 9(1): 142-152, 2020 12.
Article in English | MEDLINE | ID: mdl-32249683

ABSTRACT

Insulin action initiates a series of phosphorylation events regulating cellular differentiation, growth and metabolism. We have previously discovered, in a mass spectrometry-based phosphoproteomic study, that insulin/IGF-1 signalling induces phosphorylation of retinoid x receptor alpha (RXRα) at S22 in mouse brown pre-adipocytes. Here, we show that insulin induces the phosphorylation of RXRα at S22 in both brown precursor and mature adipocytes through a pathway involving ERK, downstream of IRS-1 and -2. We also found that RXRα S22 phosphorylation is promoted by insulin and upon re-feeding in brown adipose tissue in vivo, and that insulin-stimulated S22 phosphorylation of RXRα is dampened by diet-induced obesity. We used Rxra knockout cells re-expressing wild type (WT) or S22A non-phosphorylatable forms of RXRα to further characterize the role of S22 in brown adipocytes. Knockout of Rxra in brown pre-adipocytes resulted in decreased lipid accumulation and adipogenic gene expression during differentiation, and re-expression of RxraWT alleviated these effects. However, we observed no significant difference in cells re-expressing the RxraS22A mutant as compared with the cells re-expressing RxraWT. Furthermore, comparison of gene expression during adipogenesis in the WT and S22A re-expressing cells by RNA sequencing revealed similar transcriptomic profiles. Thus, our data propose a dispensable role for RXRα S22 phosphorylation in adipogenesis and transcription in differentiating brown pre-adipocytes.


Subject(s)
Adipocytes, Brown/metabolism , Adipogenesis , Insulin/metabolism , Retinoid X Receptor alpha/metabolism , Serine/metabolism , Adipocytes, Brown/cytology , Animals , Cell Differentiation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation
18.
Sci Transl Med ; 12(558)2020 08 26.
Article in English | MEDLINE | ID: mdl-32848096

ABSTRACT

Brown and brown-like beige/brite adipocytes dissipate energy and have been proposed as therapeutic targets to combat metabolic disorders. However, the therapeutic effects of cell-based therapy in humans remain unclear. Here, we created human brown-like (HUMBLE) cells by engineering human white preadipocytes using CRISPR-Cas9-SAM-gRNA to activate endogenous uncoupling protein 1 expression. Obese mice that received HUMBLE cell transplants showed a sustained improvement in glucose tolerance and insulin sensitivity, as well as increased energy expenditure. Mechanistically, increased arginine/nitric oxide (NO) metabolism in HUMBLE adipocytes promoted the production of NO that was carried by S-nitrosothiols and nitrite in red blood cells to activate endogenous brown fat and improved glucose homeostasis in recipient animals. Together, these data demonstrate the utility of using CRISPR-Cas9 technology to engineer human white adipocytes to display brown fat-like phenotypes and may open up cell-based therapeutic opportunities to combat obesity and diabetes.


Subject(s)
Adipocytes, Brown , Metabolic Syndrome , Adipose Tissue, Brown/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Diet, High-Fat , Energy Metabolism , Humans , Metabolic Syndrome/therapy , Mice , Mice, Obese , Obesity/metabolism , Obesity/therapy , Thermogenesis
19.
Nat Metab ; 2(5): 397-412, 2020 05.
Article in English | MEDLINE | ID: mdl-32440655

ABSTRACT

Human thermogenic adipose tissue mitigates metabolic disease, raising much interest in understanding its development and function. Here, we show that human thermogenic adipocytes specifically express a primate-specific long non-coding RNA, LINC00473 which is highly correlated with UCP1 expression and decreased in obesity and type-2 diabetes. LINC00473 is detected in progenitor cells, and increases upon differentiation and in response to cAMP. In contrast to other known adipocyte LincRNAs, LINC00473 shuttles out of the nucleus, colocalizes and can be crosslinked to mitochondrial and lipid droplet proteins. Up- or down- regulation of LINC00473 results in reciprocal alterations in lipolysis, respiration and transcription of genes associated with mitochondrial oxidative metabolism. Depletion of PLIN1 results in impaired cAMP-responsive LINC00473 expression and lipolysis, indicating bidirectional interactions between PLIN1, LINC00473 and mitochondrial oxidative functions. Thus, we suggest that LINC00473 is a key regulator of human thermogenic adipocyte function, and reveals a role for a LincRNA in inter-organelle communication and human energy metabolism.


Subject(s)
Adipocytes/physiology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/physiology , Thermogenesis/genetics , Thermogenesis/physiology , Adult , Aged , Aged, 80 and over , Cell Communication/genetics , Cell Communication/physiology , Cell Nucleus/metabolism , Cells, Cultured , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/metabolism , Energy Metabolism/genetics , Energy Metabolism/physiology , Fatty Acids, Nonesterified/metabolism , Female , Gene Expression Regulation , Humans , Lipid Droplets , Male , Middle Aged , Obesity/genetics , Obesity/metabolism , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Perilipin-1/deficiency , Perilipin-1/genetics , Uncoupling Protein 1/biosynthesis , Uncoupling Protein 1/genetics , Young Adult
20.
Nat Commun ; 11(1): 1421, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32184391

ABSTRACT

Uncoupling protein-1 (UCP1) plays a central role in energy dissipation in brown adipose tissue (BAT). Using high-throughput library screening of secreted peptides, we identify two fibroblast growth factors (FGF), FGF6 and FGF9, as potent inducers of UCP1 expression in adipocytes and preadipocytes. Surprisingly, this occurs through a mechanism independent of adipogenesis and involves FGF receptor-3 (FGFR3), prostaglandin-E2 and interaction between estrogen receptor-related alpha, flightless-1 (FLII) and leucine-rich-repeat-(in FLII)-interacting-protein-1 as a regulatory complex for UCP1 transcription. Physiologically, FGF6/9 expression in adipose is upregulated by exercise and cold in mice, and FGF9/FGFR3 expression in human neck fat is significantly associated with UCP1 expression. Loss of FGF9 impairs BAT thermogenesis. In vivo administration of FGF9 increases UCP1 expression and thermogenic capacity. Thus, FGF6 and FGF9 are adipokines that can regulate UCP1 through a transcriptional network that is dissociated from brown adipogenesis, and act to modulate systemic energy metabolism.


Subject(s)
Adipocytes, Brown/metabolism , Adipogenesis , Fibroblast Growth Factor 6/metabolism , Fibroblast Growth Factor 9/metabolism , Obesity/metabolism , Uncoupling Protein 1/metabolism , Adipocytes, Brown/cytology , Adipose Tissue, Brown/cytology , Adipose Tissue, Brown/metabolism , Animals , Fibroblast Growth Factor 6/genetics , Fibroblast Growth Factor 9/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Obesity/genetics , Obesity/physiopathology , Thermogenesis , Uncoupling Protein 1/genetics
SELECTION OF CITATIONS
SEARCH DETAIL