Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters











Publication year range
1.
Sci Adv ; 10(26): eadn5229, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38924414

ABSTRACT

There is a regional preference around lymph nodes (LNs) for adipose beiging. Here, we show that local LN removal within inguinal white adipose tissue (iWAT) greatly impairs cold-induced beiging, and this impairment can be restored by injecting M2 macrophages or macrophage-derived C-C motif chemokine (CCL22) into iWAT. CCL22 injection into iWAT effectively promotes iWAT beiging, while blocking CCL22 with antibodies can prevent it. Mechanistically, the CCL22 receptor, C-C motif chemokine receptor 4 (CCR4), within eosinophils and its downstream focal adhesion kinase/p65/interleukin-4 signaling are essential for CCL22-mediated beige adipocyte formation. Moreover, CCL22 levels are inversely correlated with body weight and fat mass in mice and humans. Acute elevation of CCL22 levels effectively prevents diet-induced body weight and fat gain by enhancing adipose beiging. Together, our data identify the CCL22-CCR4 axis as an essential mediator for LN-controlled adaptive thermogenesis and highlight its potential to combat obesity and its associated complications.


Subject(s)
Adipose Tissue, White , Chemokine CCL22 , Energy Metabolism , Lymph Nodes , Macrophages , Thermogenesis , Animals , Female , Humans , Male , Mice , Adipocytes, Beige/metabolism , Adipose Tissue, White/metabolism , Chemokine CCL22/metabolism , Eosinophils/metabolism , Lymph Nodes/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Obesity/metabolism , Receptors, CCR4/metabolism , Signal Transduction
2.
Nat Aging ; 4(6): 839-853, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38858606

ABSTRACT

Thermogenic beige adipocytes are recognized as potential therapeutic targets for combating metabolic diseases. However, the metabolic advantages that they offer are compromised with aging. Here we show that treating mice with estrogen (E2), a hormone that decreases with age, can counteract the age-related decline in beige adipogenesis when exposed to cold temperature while concurrently enhancing energy expenditure and improving glucose tolerance in mice. Mechanistically, we found that nicotinamide phosphoribosyl transferase (NAMPT) plays a pivotal role in facilitating the formation of E2-induced beige adipocytes, which subsequently suppresses the onset of age-related endoplasmic reticulum (ER) stress. Furthermore, we found that targeting NAMPT signaling, either genetically or pharmacologically, can restore the formation of beige adipocytes by increasing the number of perivascular adipocyte progenitor cells. Conversely, the absence of NAMPT signaling prevents this process. Together, our findings shed light on the mechanisms regulating the age-dependent impairment of beige adipocyte formation and underscore the E2-NAMPT-controlled ER stress pathway as a key regulator of this process.


Subject(s)
Adipocytes, Beige , Adipogenesis , Aging , Endoplasmic Reticulum Stress , Estrogens , Nicotinamide Phosphoribosyltransferase , Nicotinamide Phosphoribosyltransferase/metabolism , Animals , Adipogenesis/drug effects , Endoplasmic Reticulum Stress/drug effects , Mice , Aging/drug effects , Aging/physiology , Estrogens/metabolism , Estrogens/pharmacology , Adipocytes, Beige/drug effects , Adipocytes, Beige/metabolism , Cytokines/metabolism , Signal Transduction/drug effects , Female , Mice, Inbred C57BL , Energy Metabolism/drug effects
3.
Dev Cell ; 59(10): 1233-1251.e5, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38569546

ABSTRACT

De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively explored. Here, through in vivo lineage tracing and mouse modeling, we observed that platelet-derived growth factor receptor beta (PDGFRß)+ pericytes give rise to developmental brown adipocytes but not to those in adult homeostasis. By contrast, T-box 18 (TBX18)+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRß+ pericytes promotes brown adipogenesis by downregulating PDGFRß. Furthermore, inhibition of Notch signaling in PDGFRß+ pericytes mitigates high-fat, high-sucrose (HFHS)-induced glucose and metabolic impairment in mice during their development and juvenile phases. Collectively, these findings show that the Notch/PDGFRß axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health.


Subject(s)
Adipocytes, Brown , Adipogenesis , Cell Differentiation , Receptor, Platelet-Derived Growth Factor beta , Receptors, Notch , Stem Cells , Animals , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptors, Notch/metabolism , Mice , Adipocytes, Brown/metabolism , Adipocytes, Brown/cytology , Stem Cells/metabolism , Stem Cells/cytology , Signal Transduction , Pericytes/metabolism , Pericytes/cytology , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/cytology , Mice, Inbred C57BL , Male
4.
Mol Cell Biochem ; 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38430300

ABSTRACT

While P21-activated kinase-1 (PAK1) has been extensively studied in relation to cardiovascular health and glucose metabolism, its roles within adipose tissue and cardiometabolic diseases are less understood. In this study, we explored the effects of PAK1 deletion on energy balance, adipose tissue homeostasis, and cardiac function utilizing a whole-body PAK1 knockout (PAK1-/-) mouse model. Our findings revealed that body weight differences between PAK1-/- and WT mice emerged at 9 weeks of age, with further increases observed at 12 weeks. Furthermore, PAK1-/- mice displayed increased fat mass and decreased lean mass at 12 weeks, indicating a shift towards adiposity. In conjunction with the increased body weight, PAK1-/- mice had increased food intake and reduced energy expenditure. At a mechanistic level, PAK1 deletion boosted the expression of lipogenic markers while diminishing thermogenic markers expression in adipose tissues, contributing to reduced energy expenditure and the overall obesogenic phenotype. Moreover, our findings highlighted a significant impact on cardiac function following PAK1 deletion, including alterations in calcium kinetics and compromised systolic and lusitropy functions. In summary, our study emphasizes the significant role of PAK1 in weight regulation and cardiac function, enriching our comprehension of heart health and metabolism. These findings could potentially facilitate the identification of novel therapeutic targets in cardiometabolic diseases.

5.
Metabolism ; 151: 155740, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37995805

ABSTRACT

BACKGROUND & AIMS: Dysbiosis contributes to alcohol-associated liver disease (ALD); however, the precise mechanisms remain elusive. Given the critical role of the gut microbiota in ammonia production, we herein aim to investigate whether and how gut-derived ammonia contributes to ALD. METHODS: Blood samples were collected from human subjects with/without alcohol drinking. Mice were exposed to the Lieber-DeCarli isocaloric control or ethanol-containing diets with and without rifaximin (a nonabsorbable antibiotic clinically used for lowering gut ammonia production) supplementation for five weeks. Both in vitro (NH4Cl exposure of AML12 hepatocytes) and in vivo (urease administration for 5 days in mice) hyperammonemia models were employed. RNA sequencing and fecal amplicon sequencing were performed. Ammonia and triglyceride concentrations were measured. The gene and protein expression of enzymes involved in multiple pathways were measured. RESULTS: Chronic alcohol consumption causes hyperammonemia in both mice and human subjects. In healthy livers and hepatocytes, ammonia exposure upregulates the expression of urea cycle genes, elevates hepatic de novo lipogenesis (DNL), and increases fat accumulation. Intriguingly, ammonia promotes ethanol catabolism and acetyl-CoA formation, which, together with ammonia, synergistically facilitates intracellular fat accumulation in hepatocytes. Mechanistic investigations uncovered that ATF4 activation, as a result of ER stress induction and general control nonderepressible 2 activation, plays a central role in ammonia-provoked DNL elevation. Rifaximin ameliorates ALD pathologies in mice, concomitant with blunted hepatic ER stress induction, ATF4 activation, and DNL activation. CONCLUSIONS: An overproduction of ammonia by gut microbiota, synergistically interacting with ethanol, is a significant contributor to ALD pathologies.


Subject(s)
Ammonia , Fatty Liver , Hyperammonemia , Liver Diseases, Alcoholic , Animals , Humans , Mice , Activating Transcription Factor 4/genetics , Activating Transcription Factor 4/metabolism , Ammonia/adverse effects , Ammonia/metabolism , Ethanol/adverse effects , Ethanol/metabolism , Fatty Liver/chemically induced , Fatty Liver/metabolism , Hyperammonemia/complications , Hyperammonemia/metabolism , Hyperammonemia/pathology , Lipogenesis , Liver/metabolism , Liver Diseases, Alcoholic/metabolism , Mice, Inbred C57BL , Rifaximin/pharmacology
6.
bioRxiv ; 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37693431

ABSTRACT

Thermogenic beige adipocytes are recognized as potential therapeutic targets for combating metabolic diseases. However, the metabolic advantages they offer are compromised with aging. Here, we show that treating mice with estrogen (E2), a hormone that decreases with age, to mice can counteract the aging- related decline in beige adipocyte formation when subjected to cold, while concurrently enhancing energy expenditure and improving glucose tolerance. Mechanistically, we find that nicotinamide phosphoribosyltranferase (NAMPT) plays a pivotal role in facilitating the formation of E2-induced beige adipocytes, which subsequently suppresses the onset of age-related ER stress. Furthermore, we found that targeting NAMPT signaling, either genetically or pharmacologically, can restore the formation of beige adipocytes by increasing the number of perivascular adipocyte progenitor cells. Conversely, the absence of NAMPT signaling prevents this process. In conclusion, our findings shed light on the mechanisms governing the age-dependent impairment of beige adipocyte formation and underscore the E2-NAMPT controlled ER stress as a key regulator of this process. Highlights: Estrogen restores beige adipocyte failure along with improved energy metabolism in old mice.Estrogen enhances the thermogenic gene program by mitigating age-induced ER stress.Estrogen enhances the beige adipogenesis derived from SMA+ APCs.Inhibiting the NAMPT signaling pathway abolishes estrogen-promoted beige adipogenesis.

7.
bioRxiv ; 2023 May 24.
Article in English | MEDLINE | ID: mdl-37293108

ABSTRACT

De novo brown adipogenesis holds potential in combating the epidemics of obesity and diabetes. However, the identity of brown adipocyte progenitor cells (APCs) and their regulation have not been extensively studied. Here through in vivo lineage tracing, we observed that PDGFRß+ pericytes give rise to developmental brown adipocytes, but not to those in adult homeostasis. In contrast, TBX18+ pericytes contribute to brown adipogenesis throughout both developmental and adult stages, though in a depot-specific manner. Mechanistically, Notch inhibition in PDGFRß+ pericytes promotes brown adipogenesis through the downregulation of PDGFRß. Furthermore, inhibition of Notch signaling in PDGFRß+ pericytes mitigates HFHS (high-fat, high-sucrose) induced glucose and metabolic impairment in both developmental and adult stages. Collectively, these findings show that the Notch/PDGFRß axis negatively regulates developmental brown adipogenesis, and its repression promotes brown adipose tissue expansion and improves metabolic health. Highlights: PDGFRß+ pericytes act as an essential developmental brown APC.TBX18+ pericytes contribute to brown adipogenesis in a depot-specific manner.Inhibiting Notch-Pdgfrß axis promotes brown APC adipogenesis.Enhanced postnatal brown adipogenesis improves metabolic health in adult stage.

8.
Nat Commun ; 14(1): 2731, 2023 05 12.
Article in English | MEDLINE | ID: mdl-37169793

ABSTRACT

A potential therapeutic target to curb obesity and diabetes is thermogenic beige adipocytes. However, beige adipocytes quickly transition into white adipocytes upon removing stimuli. Here, we define the critical role of cyclin dependent kinase inhibitor 2A (Cdkn2a) as a molecular pedal for the beige-to-white transition. Beige adipocytes lacking Cdkn2a exhibit prolonged lifespan, and male mice confer long-term metabolic protection from diet-induced obesity, along with enhanced energy expenditure and improved glucose tolerance. Mechanistically, Cdkn2a promotes the expression and activity of beclin 1 (BECN1) by directly binding to its mRNA and its negative regulator BCL2 like 1 (BCL2L1), activating autophagy and accelerating the beige-to-white transition. Reactivating autophagy by pharmacological or genetic methods abolishes beige adipocyte maintenance induced by Cdkn2a ablation. Furthermore, hyperactive BECN1 alone accelerates the beige-to-white transition in mice and human. Notably, both Cdkn2a and Becn1 exhibit striking positive correlations with adiposity. Hence, blocking Cdkn2a-mediated BECN1 activity holds therapeutic potential to sustain beige adipocytes in treating obesity and related metabolic diseases.


Subject(s)
Adipocytes, Beige , Adipose Tissue, Beige , Obesity , Animals , Humans , Male , Mice , Adipocytes, Beige/metabolism , Adipocytes, White/metabolism , Adipose Tissue, Beige/metabolism , Adipose Tissue, White/metabolism , Adiposity/genetics , Adiposity/physiology , Obesity/genetics , Obesity/metabolism , Thermogenesis
9.
Hepatol Commun ; 7(3): e0059, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36757400

ABSTRACT

BACKGROUND: Adipose tissue thermogenic activities use fatty acids from lipolysis for heat generation. Therefore, a tight coupling between lipolysis and thermogenesis is physiologically imperative in maintaining not only body temperature but also lipids homeostasis. Adipose tissue dysfunction contributes to alcoholic liver disease (ALD). Here, studies were conducted to examine how alcohol intake affects adipose tissue thermogenic activities and whether altered adipose tissue thermogenesis contributes to ALD. METHODS: Both the Lieber-DeCarli and the NIAAA mouse models of ALD were used. Denervation surgery in epididymal fat pads was performed. CL316,243, a selective ß3-adrenoceptor agonist, SR59230A, a selective ß3 adrenoceptor (ADRB3) antagonist, and rapamycin, a selective mechanistic target of rapamycin complex 1 (mTORC1) inhibitor, were administrated through i.p. injection. Adipocyte-specific Prdm16 knockout mice were subjected to alcohol-containing diet chronically. RESULTS: Chronic alcohol consumption, which enhances adipose tissue lipolysis, inhibits thermogenic activities of beige adipocytes in inguinal white adipose tissue (WAT), leading to an uncoupling status between lipolysis and thermogenesis in WAT at both basal and ADRB3 stimulation states. CL316,243 administration exacerbates liver pathologies of ALD. Alcohol intake inhibits mTORC1 activities in WAT. In mice, mTORC1 inhibition by rapamycin inhibits the thermogenesis of iWAT, whereas enhancing WAT lipolysis. Further investigations using adipocyte-specific Prdm16 knockout mice revealed that functional deficiency of beige adipocytes aggravates liver pathologies of ALD, suggesting that the inhibitory effect of alcohol on WAT browning/thermogenesis contributes to ALD pathogenesis. CONCLUSION: Chronic alcohol consumption induces an "uncoupling status" between lipolysis and browning/thermogenesis in WAT by inhibiting mTORC1 activation. Diminished WAT browning/thermogenesis, concomitant with enhanced lipolysis, contributes to ALD pathogenesis.


Subject(s)
Lipolysis , Liver Diseases, Alcoholic , Mice , Animals , Lipolysis/physiology , Adipose Tissue, Brown/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Adipose Tissue, White/metabolism , Thermogenesis/physiology , Liver Diseases, Alcoholic/metabolism , Mice, Knockout , Receptors, Adrenergic/metabolism
10.
Am J Physiol Cell Physiol ; 321(3): C585-C595, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34378991

ABSTRACT

Defined as the dysfunction and/or cell death caused by toxic lipids accumulation in hepatocytes, hepatic lipotoxicity plays a pathological role in nonalcoholic fatty liver disease. The cellular and molecular mechanisms underlying lipotoxicity remain to be elucidated. In this study, using AML12 cells, a nontransformed murine hepatocyte cell line, exposed to palmitate (a 16-C saturated fatty acid) as an experimental model, we investigated the role and mechanisms of nicotinamide N-methyltransferase (NNMT), a methyltransferase catalyzing nicotinamide methylation and degradation, in hepatic lipotoxicity. We initially identified activating transcription factor 4 (ATF4) as a major transcription factor for hepatic NNMT expression. Here, we demonstrated that palmitate upregulates NNMT expression via activating ATF4 in a mechanistic target of rapamycin complex 1 (mTORC1)-dependent mechanism in that mTORC1 inhibition by both Torin1 and rapamycin attenuated ATF4 activation and NNMT upregulation. We further demonstrated that the mTORC1-dependent ATF4 activation is an integral signaling event of unfolded protein response (UPR) as both ATF4 activation and NNMT upregulation by tunicamycin, a well-documented endoplasmic reticulum (ER) stress inducer, are blunted when hepatocytes were pretreated with Torin1. Importantly, our data uncovered that NNMT upregulation contributes to palmitate-induced hepatotoxicity as NNMT inhibition, via either pharmacological (NNMT inhibitors) or genetic approach (siRNA transfection), provided protection against palmitate lipotoxicity. Our further mechanistic exploration identified protein kinase A (PKA) activation to contribute, at least, partially to the protective effect of NNMT inhibition against lipotoxicity. Collectively, our data demonstrated that NNMT upregulation by the mTORC1-ATF4 pathway activation contributes to the development of lipotoxicity in hepatocytes.


Subject(s)
Activating Transcription Factor 4/metabolism , Hepatocytes/drug effects , Mechanistic Target of Rapamycin Complex 1/metabolism , Nicotinamide N-Methyltransferase/metabolism , Palmitates/toxicity , Activating Transcription Factor 4/genetics , Animals , Cell Death/drug effects , Cell Line , Cyclic AMP-Dependent Protein Kinases/metabolism , Endoplasmic Reticulum Stress/drug effects , Hepatocytes/enzymology , Hepatocytes/pathology , Male , Mice , Mice, Inbred C57BL , Nicotinamide N-Methyltransferase/genetics , Signal Transduction , Unfolded Protein Response/drug effects , Up-Regulation , X-Box Binding Protein 1/genetics , X-Box Binding Protein 1/metabolism
11.
Dev Cell ; 56(7): 985-999.e4, 2021 04 05.
Article in English | MEDLINE | ID: mdl-33711247

ABSTRACT

Thermogenic beige fat found in white adipose tissue is a potential therapeutic target to curb the global obesity and diabetes epidemic. However, these inducible thermogenic beige adipocytes have been thought to be short-lived and to rapidly convert to "white-like" adipocytes after discontinuing stimuli. In this study, using effective labeling techniques and genetic mouse tools, we demonstrate that a subset of UCP1+ cells that exist within white adipose tissue are able to self-divide and contribute to new beige adipocyte recruitment in response to ß3 stimuli. When these cells are depleted or their adipogenic capability is blocked, ß3-induced beige adipocyte formation is impaired. We also identify a cell-cycle machinery of p21 and CDKN2A as a molecular basis of beige adipocyte regulation. Collectively, our findings provide new insights into the cellular and molecular mechanisms of beige adipocyte regulation and potential therapeutic opportunities to induce the beige phenotype and treat metabolic disease.


Subject(s)
Adipocytes, Beige/physiology , Adipose Tissue, White/cytology , Stem Cells/physiology , Uncoupling Protein 1/analysis , Adrenergic beta-3 Receptor Agonists/pharmacology , Animals , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Gene Deletion , Genes, cdc , Male , Mice , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism
12.
Elife ; 92020 06 19.
Article in English | MEDLINE | ID: mdl-32553115

ABSTRACT

Adipocytes arise from distinct progenitor populations during developmental and adult stages but little is known about how developmental progenitors differ from adult progenitors. Here, we investigate the role of platelet-derived growth factor receptor alpha (PDGFRα) in the divergent regulation of the two different adipose progenitor cells (APCs). Using in vivo adipose lineage tracking and deletion mouse models, we found that developmental PDGFRα+ cells are adipogenic and differentiated into mature adipocytes, and the deletion of Pdgfra in developmental adipose lineage disrupted white adipose tissue (WAT) formation. Interestingly, adult PDGFRα+ cells do not significantly contribute to adult adipogenesis, and deleting Pdgfra in adult adipose lineage did not affect WAT homeostasis. Mechanistically, embryonic APCs require PDGFRα for fate maintenance, and without PDGFRα, they underwent fate change from adipogenic to fibrotic lineage. Collectively, our findings indicate that PDGFRα+ cells and Pdgfra gene itself are differentially required for WAT development and adult WAT homeostasis.


Subject(s)
Adipogenesis/genetics , Adipose Tissue/growth & development , Homeostasis , Receptor, Platelet-Derived Growth Factor alpha/genetics , Stem Cells/metabolism , Adipose Tissue/metabolism , Adipose Tissue, White/growth & development , Adipose Tissue, White/metabolism , Animals , Cell Differentiation , Male , Mice , Mice, Transgenic , Receptor, Platelet-Derived Growth Factor alpha/metabolism
13.
Biochim Biophys Acta Gene Regul Mech ; 1862(6): 643-656, 2019 06.
Article in English | MEDLINE | ID: mdl-30959128

ABSTRACT

Gluconeogenesis is essential for blood glucose homeostasis during fasting and is regulated by various enzymes, which are encoded by gluconeogenic genes. Those genes are controlled by various transcription factors. Zinc finger and BTB domain-containing 7c (Zbtb7c, also called Kr-pok) is a BTB-POZ family transcription factor with proto-oncogenic activity. Previous findings have indicated that Zbtb7c is involved in the regulation of fatty acid biosynthesis, suggesting an involvement also in primary metabolism. We found here that fasting induced Zbtb7c expression in the mouse liver and in primary liver hepatocytes. We also observed that Zbtb7c-knockout mice have decreased blood glucose levels, so we investigated whether Zbtb7c plays a role in gluconeogenesis. Indeed, differential gene expression analysis of Zbtb7c-knockout versus wild type mouse livers showed downregulated transcription of gluconeogenic genes encoding the glucose 6-phosphatase catalytic subunit (G6pc) and phosphoenolpyruvate carboxykinase 1 (Pck1), while Zbtb7c expression upregulated these two genes, under fasting conditions. Mechanistically, we found that when complexed with histone deacetylase 3 (Hdac3), Zbtb7c binds insulin response elements (IREs) within the G6pc and Pck1 promoters. Moreover, complexed Zbtb7c deacetylated forkhead box O1 (Foxo1), thereby increasing Foxo1 binding to the G6pc and Pck1 IREs, resulting in their transcriptional activation. These results demonstrate Zbtb7c to be a crucial metabolic regulator of blood glucose homeostasis, during mammalian fasting.


Subject(s)
Fasting , Gene Expression Regulation , Gluconeogenesis/physiology , Glucose-6-Phosphatase/genetics , Intracellular Signaling Peptides and Proteins/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Proteins/metabolism , Transcription Factors/metabolism , Zinc Fingers/physiology , Animals , Blood Glucose , DNA-Binding Proteins/metabolism , Fatty Acids/biosynthesis , Forkhead Box Protein O1/metabolism , Gluconeogenesis/genetics , Glucose/metabolism , Glucose-6-Phosphatase/metabolism , HEK293 Cells , Hep G2 Cells , Hepatocytes/metabolism , Histone Deacetylases/metabolism , Homeostasis , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Mutagenesis, Site-Directed , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , Promoter Regions, Genetic , Proteins/genetics , Transcriptome , Zinc Fingers/genetics
14.
Biochim Biophys Acta Mol Basis Dis ; 1864(6 Pt A): 2097-2107, 2018 06.
Article in English | MEDLINE | ID: mdl-29601978

ABSTRACT

Prolactin regulatory element-binding (PREB) protein is a transcription factor that regulates prolactin (PRL) gene expression. PRL, also known as luteotropic hormone or luteotropin, is well known for its role in producing milk. However, the role of PREB, in terms of hepatic glucose metabolism, is not well elucidated. Here, we observed expression of Preb in the mouse liver, in connection with glucose homeostasis. Morevoer, Preb was downregulated in db/db, ob/ob and high-fat diet-induced obese (DIO) mice, concurrent with upregulation of the liver genes glucose-6-phosphatase (G6pc) and phosphoenolpyruvate carboxykinase-1 (Pck). Administration of adenovirus-Preb (Ad-Preb) to db/db, ob/ob, and DIO mice diminished glucose, insulin, and pyruvate tolerance, which analogously, were impaired in normal (C57BL/6) mice knocked down for Preb, via infection with Ad-shPreb (anti-Preb RNA), indicating Preb to be a negative regulator of liver gluconeogenic genes. We further demonstrate that Preb negatively influences gluconeogenic gene expression, by directly binding to their promoters at a prolactin core-binding element (PCBE). A better understanding of Preb gene expression, during the pathogenesis of hepatic insulin resistance, could ultimately provide new avenues for therapies for metabolic syndrome, obesity, and type-2 diabetes mellitus, disorders whose worldwide incidences are increasing drastically.


Subject(s)
DNA-Binding Proteins/metabolism , Gluconeogenesis , Glucose/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Liver/metabolism , Transcription Factors/metabolism , Animals , Blood Glucose , DNA-Binding Proteins/genetics , Diet, High-Fat/adverse effects , Disease Models, Animal , Down-Regulation , Fasting , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Hepatocytes/metabolism , Humans , Insulin/metabolism , Liver/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/blood , Obesity/etiology , Obesity/metabolism , Primary Cell Culture , Prolactin/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transcription Factors/genetics , Up-Regulation
15.
Sci Rep ; 7: 45300, 2017 03 24.
Article in English | MEDLINE | ID: mdl-28338058

ABSTRACT

The incidence of prostate cancer (PC) is growing rapidly throughout the world, in probable association with the adoption of western style diets. Thus, understanding the molecular pathways triggering the development of PC is crucial for both its prevention and treatment. Here, we investigated the role of the metabolism-associated protein, CREB3L4, in the proliferation of PC cells. CREB3L4 was upregulated by the synthetic androgen, R1881, in LNCaP PC cells (an androgen-dependent cell line). Knockdown of CREB3L4 resulted in decreased androgen-dependent PC cell growth. LNCaP cells transfected with siCREB3L4 underwent G2/M arrest, with upregulation of the proteins cyclin B1, phospho-CDK1, p21Waf1/Cip1, and INCA1, and downregulation of cyclin D1. Moreover, depletion of CREB3L4 resulted in significantly decreased expression of a subset of androgen-receptor (AR) target genes, including PSA, FKBP5, HPGD, KLK2, and KLK4. We also demonstrated that CREB3L4 directly interacts with the AR, and increases the binding of AR to androgen response elements (AREs). We also identified a role for the unfolded protein response (and its surrogate, IRE1α), in activating CREB3L4. Cumulatively, we postulate that CREB3L4 expression is mediated by an AR-IRE1α axis, but is also directly regulated by AR-to-ARE binding. Thus, our study demonstrates that CREB3L4 plays a key role in PC cell proliferation, which is promoted by both AR and IRE1α.


Subject(s)
Basic-Leucine Zipper Transcription Factors/metabolism , Cell Proliferation , Nuclear Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein , Down-Regulation/drug effects , Endoribonucleases/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Male , Metribolone/pharmacology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Protein Binding , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Unfolded Protein Response/drug effects , Up-Regulation/drug effects
16.
Sci Rep ; 5: 17395, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26620281

ABSTRACT

Glucokinase (GK), mainly expressed in the liver and pancreatic ß-cells, is critical for maintaining glucose homeostasis. GK expression and kinase activity, respectively, are both modulated at the transcriptional and post-translational levels. Post-translationally, GK is regulated by binding the glucokinase regulatory protein (GKRP), resulting in GK retention in the nucleus and its inability to participate in cytosolic glycolysis. Although hepatic GKRP is known to be regulated by allosteric mechanisms, the precise details of modulation of GKRP activity, by post-translational modification, are not well known. Here, we demonstrate that GKRP is acetylated at Lys5 by the acetyltransferase p300. Acetylated GKRP is resistant to degradation by the ubiquitin-dependent proteasome pathway, suggesting that acetylation increases GKRP stability and binding to GK, further inhibiting GK nuclear export. Deacetylation of GKRP is effected by the NAD(+)-dependent, class III histone deacetylase SIRT2, which is inhibited by nicotinamide. Moreover, the livers of db/db obese, diabetic mice also show elevated GKRP acetylation, suggesting a broader, critical role in regulating blood glucose. Given that acetylated GKRP may affiliate with type-2 diabetes mellitus (T2DM), understanding the mechanism of GKRP acetylation in the liver could reveal novel targets within the GK-GKRP pathway, for treating T2DM and other metabolic pathologies.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Carrier Proteins/metabolism , Diabetes Mellitus, Type 2/metabolism , Glucokinase/metabolism , Glucose/metabolism , Protein Processing, Post-Translational , Acetylation , Adaptor Proteins, Signal Transducing/genetics , Animals , Carrier Proteins/genetics , Diabetes Mellitus, Type 2/genetics , Glucokinase/genetics , Glucose/genetics , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Male , Mice , Mice, Obese , Sirtuin 2/genetics , Sirtuin 2/metabolism
17.
Protein Cell ; 6(11): 804-13, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26334401

ABSTRACT

Post-translational modifications (PTMs) of transcription factors play a crucial role in regulating metabolic homeostasis. These modifications include phosphorylation, methylation, acetylation, ubiquitination, SUMOylation, and O-GlcNAcylation. Recent studies have shed light on the importance of lysine acetylation at nonhistone proteins including transcription factors. Acetylation of transcription factors affects subcellular distribution, DNA affinity, stability, transcriptional activity, and current investigations are aiming to further expand our understanding of the role of lysine acetylation of transcription factors. In this review, we summarize recent studies that provide new insights into the role of protein lysine-acetylation in the transcriptional regulation of metabolic homeostasis.


Subject(s)
Transcription Factors/metabolism , Acetylation , Animals , Diabetes Mellitus, Type 2/metabolism , Homeostasis/genetics , Homeostasis/physiology , Humans , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology
18.
Yonsei Med J ; 54(3): 545-59, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23549795

ABSTRACT

Peroxisome proliferator-activated receptor gamma (PPARγ) belongs to a nuclear receptor superfamily; members of which play key roles in the control of body metabolism principally by acting on adipose tissue. Ligands of PPARγ, such as thiazolidinediones, are widely used in the treatment of metabolic syndromes and type 2 diabetes mellitus (T2DM). Although these drugs have potential benefits in the treatment of T2DM, they also cause unwanted side effects. Thus, understanding the molecular mechanisms governing the transcriptional activity of PPARγ is of prime importance in the development of new selective drugs or drugs with fewer side effects. Recent advancements in molecular biology have made it possible to obtain a deeper understanding of the role of PPARγ in body homeostasis. The transcriptional activity of PPARγ is subject to regulation either by interacting proteins or by modification of the protein itself. New interacting partners of PPARγ with new functions are being unveiled. In addition, post-translational modification by various cellular signals contributes to fine-tuning of the transcriptional activities of PPARγ. In this review, we will summarize recent advancements in our understanding of the post-translational modifications of, and proteins interacting with, PPARγ, both of which affect its transcriptional activities in relation to adipogenesis.


Subject(s)
Models, Genetic , PPAR gamma/physiology , Protein Processing, Post-Translational , Gene Expression Regulation , Homeostasis , PPAR gamma/genetics , PPAR gamma/metabolism , Sumoylation , Transcription Factors/metabolism , Transcription Factors/physiology , Ubiquitination
19.
Exp Diabetes Res ; 2012: 716425, 2012.
Article in English | MEDLINE | ID: mdl-22110478

ABSTRACT

Non-alcoholic fatty liver disease (NAFLD) is characterized by fat accumulation in the liver not due to alcohol abuse. NAFLD is accompanied by variety of symptoms related to metabolic syndrome. Although the metabolic link between NAFLD and insulin resistance is not fully understood, it is clear that NAFLD is one of the main cause of insulin resistance. NAFLD is shown to affect the functions of other organs, including pancreas, adipose tissue, muscle and inflammatory systems. Currently efforts are being made to understand molecular mechanism of interrelationship between NAFLD and insulin resistance at the transcriptional level with specific focus on post-translational modification (PTM) of transcription factors. PTM of transcription factors plays a key role in controlling numerous biological events, including cellular energy metabolism, cell-cycle progression, and organ development. Cell type- and tissue-specific reversible modifications include lysine acetylation, methylation, ubiquitination, and SUMOylation. Moreover, phosphorylation and O-GlcNAcylation on serine and threonine residues have been shown to affect protein stability, subcellular distribution, DNA-binding affinity, and transcriptional activity. PTMs of transcription factors involved in insulin-sensitive tissues confer specific adaptive mechanisms in response to internal or external stimuli. Our understanding of the interplay between these modifications and their effects on transcriptional regulation is growing. Here, we summarize the diverse roles of PTMs in insulin-sensitive tissues and their involvement in the pathogenesis of insulin resistance.


Subject(s)
Fatty Liver/physiopathology , Insulin Resistance/physiology , Protein Processing, Post-Translational , Transcription Factors/metabolism , Adipokines/physiology , Animals , DNA/metabolism , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/physiopathology , Fatty Acids, Nonesterified/physiology , Fatty Liver/complications , Humans , Insulin-Secreting Cells/physiology , Liver/metabolism , Macrophages/physiology , Metabolic Syndrome/complications , Metabolic Syndrome/physiopathology , Non-alcoholic Fatty Liver Disease
20.
Biochem Biophys Res Commun ; 403(3-4): 329-34, 2010 Dec 17.
Article in English | MEDLINE | ID: mdl-21078299

ABSTRACT

During a state of fasting, the blood glucose level is maintained by hepatic gluconeogenesis. SIRT1 is an important metabolic regulator during nutrient deprivation and the liver-specific knockdown of SIRT1 resulted in decreased glucose production. We hypothesize that SIRT1 is responsible for the upregulation of insulin-suppressed gluconeogenic genes through the deacetylation of FOXO1. Treatment of primary cultured hepatocytes with resveratrol increased insulin-repressed PEPCK and G6Pase mRNA levels, which depend on SIRT1 activity. We found that the resveratrol treatment resulted in a decrease in the phosphorylation of Akt and FOXO1, which are independent of SIRT1 action. Fluorescence microscopy revealed that resveratrol caused the nuclear localization of FOXO1. In the nucleus, FOXO1 is deacetylated by SIRT1, which might make it more accessible to the IRE of the PEPCK and G6Pase promoter, causing an increase in their gene expression. Our results indicate that resveratrol upregulates the expression of gluconeogenic genes by attenuating insulin signaling and by deacetylating FOXO1, which are SIRT1-independent in the cytosol and SIRT1-dependent in the nucleus, respectively.


Subject(s)
Antioxidants/pharmacology , Forkhead Transcription Factors/metabolism , Gene Expression/drug effects , Gluconeogenesis/drug effects , Liver/drug effects , Stilbenes/pharmacology , Animals , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gluconeogenesis/genetics , Humans , Liver/metabolism , Phosphorylation , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Resveratrol , Sirtuin 1/genetics , Sirtuin 1/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL