1
|
Li YJ, Zhang C, Martincuks A, Herrmann A, Yu H. STAT proteins in cancer: orchestration of metabolism. Nat Rev Cancer 2023; 23:115-134. [PMID: 36596870 DOI: 10.1038/s41568-022-00537-3] [Citation(s) in RCA: 106] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2022] [Indexed: 01/04/2023]
Abstract
Reprogrammed metabolism is a hallmark of cancer. However, the metabolic dependency of cancer, from tumour initiation through disease progression and therapy resistance, requires a spectrum of distinct reprogrammed cellular metabolic pathways. These pathways include aerobic glycolysis, oxidative phosphorylation, reactive oxygen species generation, de novo lipid synthesis, fatty acid β-oxidation, amino acid (notably glutamine) metabolism and mitochondrial metabolism. This Review highlights the central roles of signal transducer and activator of transcription (STAT) proteins, notably STAT3, STAT5, STAT6 and STAT1, in orchestrating the highly dynamic metabolism not only of cancer cells but also of immune cells and adipocytes in the tumour microenvironment. STAT proteins are able to shape distinct metabolic processes that regulate tumour progression and therapy resistance by transducing signals from metabolites, cytokines, growth factors and their receptors; defining genetic programmes that regulate a wide range of molecules involved in orchestration of metabolism in cancer and immune cells; and regulating mitochondrial activity at multiple levels, including energy metabolism and lipid-mediated mitochondrial integrity. Given the central role of STAT proteins in regulation of metabolic states, they are potential therapeutic targets for altering metabolic reprogramming in cancer.
Collapse
Affiliation(s)
- Yi-Jia Li
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Chunyan Zhang
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Antons Martincuks
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
| | - Andreas Herrmann
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA
- Sorrento Therapeutics, San Diego, CA, USA
| | - Hua Yu
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, USA.
| |
Collapse
|
2
|
Anhê GF, Bordin S. The adaptation of maternal energy metabolism to lactation and its underlying mechanisms. Mol Cell Endocrinol 2022; 553:111697. [PMID: 35690287 DOI: 10.1016/j.mce.2022.111697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 04/15/2022] [Accepted: 06/01/2022] [Indexed: 11/29/2022]
Abstract
Maternal energy metabolism undergoes a singular adaptation during lactation that allows for the caloric enrichment of milk. Changes in the mammary gland, changes in the white adipose tissue, brown adipose tissue, liver, skeletal muscles and endocrine pancreas are pivotal for this adaptation. The present review details the landmark studies describing the enzymatic modulation and the endocrine signals behind these metabolic changes. We will also update this perspective with data from recent studies showing transcriptional and post-transcriptional mechanisms that mediate the adaptation of the maternal metabolism to lactation. The present text will also bring experimental and observational data that describe the long-term consequences that short periods of lactation impose to maternal metabolism.
Collapse
Affiliation(s)
- Gabriel Forato Anhê
- Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas, Brazil.
| | - Silvana Bordin
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
3
|
Harlan B, Park HG, Spektor R, Cummings B, Brenna JT, Soloway PD. Single-cell chromatin accessibility and lipid profiling reveals SCD1-dependent metabolic shift in adipocytes induced by bariatric surgery. PLoS One 2021; 16:e0261783. [PMID: 34972124 PMCID: PMC8719700 DOI: 10.1371/journal.pone.0261783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/09/2021] [Indexed: 11/23/2022] Open
Abstract
Obesity promotes type 2 diabetes and cardiometabolic pathologies. Vertical sleeve gastrectomy (VSG) is used to treat obesity resulting in long-term weight loss and health improvements that precede weight loss; however, the mechanisms underlying the immediate benefits remain incompletely understood. Because adipose plays a crucial role in energy homeostasis and utilization, we hypothesized that VSG exerts its influences, in part, by modulating adipose functional states. We applied single-cell ATAC sequencing and lipid profiling to inguinal and epididymal adipose depots from mice that received sham surgery or VSG. We observed depot-specific cellular composition and chromatin accessibility patterns that were altered by VSG. Specifically, accessibility at Scd1, a fatty acid desaturase, was substantially reduced after VSG in mature adipocytes of inguinal but not epididymal depots. This was accompanied by reduced accumulation of SCD1-produced unsaturated fatty acids. Given these findings and reports that reductions in Scd1 attenuate obesity and insulin resistance our results suggest VSG exerts its beneficial effects through an inguinal depot-specific reduction of SCD1 activity.
Collapse
Affiliation(s)
- Blaine Harlan
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Hui Gyu Park
- Dell Pediatric Research Institute, Department of Pediatrics, University of Texas at Austin, Austin, Texas, United States of America
| | - Roman Spektor
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
| | - Bethany Cummings
- Department of Surgery, School of Medicine, University of California, Davis, Sacramento, California, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - J. Thomas Brenna
- Dell Pediatric Research Institute, Department of Pediatrics, University of Texas at Austin, Austin, Texas, United States of America
- Division of Nutritional Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States of America
| | - Paul D. Soloway
- Field of Genetics, Genomics, and Development, Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York, United States of America
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
- Division of Nutritional Sciences, College of Agriculture and Life Sciences, Cornell University, Ithaca, New York, United States of America
| |
Collapse
|
4
|
Lourido F, Quenti D, Salgado-Canales D, Tobar N. Domeless receptor loss in fat body tissue reverts insulin resistance induced by a high-sugar diet in Drosophila melanogaster. Sci Rep 2021; 11:3263. [PMID: 33547367 PMCID: PMC7864986 DOI: 10.1038/s41598-021-82944-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 01/27/2021] [Indexed: 02/07/2023] Open
Abstract
Insulin resistance is a hallmark of type 2 diabetes resulting from the confluence of several factors, including genetic susceptibility, inflammation, and diet. Under this pathophysiological condition, the dysfunction of the adipose tissue triggered by the excess caloric supply promotes the loss of sensitivity to insulin at the local and peripheral level, a process in which different signaling pathways are involved that are part of the metabolic response to the diet. Besides, the dysregulation of insulin signaling is strongly associated with inflammatory processes in which the JAK/STAT pathway plays a central role. To better understand the role of JAK/STAT signaling in the development of insulin resistance, we used a simple organism, Drosophila melanogaster, as a type 2 diabetes model generated by the consumption of a high-sugar diet. In this model, we studied the effects of inhibiting the expression of the JAK/STAT pathway receptor Domeless, in fat body, on adipose metabolism and glycemic control. Our results show that the Domeless receptor loss in fat body cells reverses both hyperglycemia and the increase in the expression of the insulin resistance marker Nlaz, observed in larvae fed a high sugar diet. This effect is consistent with a significant reduction in Dilp2 mRNA expression and an increase in body weight compared to wild-type flies fed high sugar diets. Additionally, the loss of Domeless reduced the accumulation of triglycerides in the fat body cells of larvae fed HSD and also significantly increased the lifespan of adult flies. Taken together, our results show that the loss of Domeless in the fat body reverses at least in part the dysmetabolism induced by a high sugar diet in a Drosophila type 2 diabetes model.
Collapse
Affiliation(s)
- Fernanda Lourido
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile
| | - Daniela Quenti
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile
| | - Daniela Salgado-Canales
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile
| | - Nicolás Tobar
- Cellular Biology Laboratory, Institute of Nutrition and Food Technology (INTA), University of Chile, Av. El Líbano, 5524, Macul, Santiago, Chile.
| |
Collapse
|
5
|
Zhang Y, Zhou L, Zhang Z, Xu Q, Han X, Zhao Y, Song X, Zhao T, Ye L. Effects of di (2-ethylhexyl) phthalate and high-fat diet on lipid metabolism in rats by JAK2/STAT5. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2020; 27:3837-3848. [PMID: 31732953 DOI: 10.1007/s11356-019-06599-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 09/24/2019] [Indexed: 06/10/2023]
Abstract
Exposure to di (2-ethylhexyl) phthalate (DEHP) induces lipid metabolism disorder and high-fat diet (HD) may have joint effects with DEHP. We aim to clarify the role of JAK2/STAT5 pathway in the process and reveal the effects of HD on the toxicity of DEHP. Wistar rats (160 animals) were fed with HD or normal diet (ND) respectively and exposed to DEHP 0, 5, 50, and 500 mg/kg/day for 8 weeks. Lipid levels, as well as the morphology of liver and adipose, mRNA levels, and protein levels of JAK2, STAT5A, STAT5B, FAS, ap2, and PDK4 were detected. The results showed that DEHP exposure leads to increased weight gain. The JAK2/STAT5 pathway was activated in adipose after DEHP exposure and promoted the expression of FAS, ap2, and PDK4 in ND rats. While in the liver, JAK2 was inhibited, and lipid synthesis and accumulation were increased. However, rats exposed to DEHP in combination with HD showed a complete disorder of lipid metabolism. Therefore, we conclude that DEHP affects lipid metabolism through regulating the JAK2/STAT5 pathway and promotes adipogenesis and lipid accumulation. High-fat diet may have a joint effect with DEHP on lipid metabolism disorder.
Collapse
Affiliation(s)
- Yuezhu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Zhaoming Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Qi Xu
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Xu Han
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Yaming Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Xinyue Song
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Tianyang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, 1163 Xin Min Street, Changchun, 130021, China.
| |
Collapse
|
6
|
Zhang Y, Wang S, Zhao T, Yang L, Guo S, Shi Y, Zhang X, Zhou L, Ye L. Mono-2-ethylhexyl phthalate (MEHP) promoted lipid accumulation via JAK2/STAT5 and aggravated oxidative stress in BRL-3A cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2019; 184:109611. [PMID: 31491605 DOI: 10.1016/j.ecoenv.2019.109611] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 08/21/2019] [Accepted: 08/24/2019] [Indexed: 06/10/2023]
Abstract
Mono-2-ethylhexyl phthalate (MEHP), as the major metabolite of Di-(2-ethylhexyl) phthalate (DEHP), can induce lipid accumulation in hepatocytes and further leads to non-alcoholic fatty liver disease (NAFLD), while the underlying mechanism is unclear. We aim to clarify the effects of JAK2/STAT5 pathway on lipid accumulation induced by MEHP and the role of oxidation stress in NAFLD. BRL-3A hepatocytes were exposed to MEHP (0, 10, 50, 100 and 200 μM) for 24 h and 48 h. Then the lipid droplets in cells were observed by Oil-Red-O staining and quantified by isopropyl alcohol. The levels of TG, SOD, TBARS, AST and ALT were all detected by commercial kits. RT-PCR was used to detect mRNA expression, and western blotting was used to detect the expression of proteins encoded by JAK2/STAT5 pathway genes and lipid metabolism-related genes. As a result, MEHP promoted the lipid synthesis and accumulation in BRL-3A cells. MEHP down-regulated the expression and inhibited the activation of JAK2/STAT5. Moreover, the lipid metabolism-related kinases levels were elevated after MEHP exposure. In addition, the SOD levels were gradually decreased and the TBARS levels were increased in MEHP-treated groups. The lipid metabolism-related proteins levels were correlated with the oxidation stress levels. Furthermore, the ALT and AST levels were elevated after MEHP exposure. Therefore, we concluded that MEHP led to lipid accumulation through inhibiting JAK2/STAT5 pathway, resulted in damaging liver parenchyma and NAFLD by aggravating oxidation stress.
Collapse
Affiliation(s)
- Yuezhu Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Shuyue Wang
- Department of Emergency, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Tianyang Zhao
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Liwei Yang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Shuangyu Guo
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Yanbin Shi
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Xueting Zhang
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China
| | - Liting Zhou
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China.
| | - Lin Ye
- Department of Occupational and Environmental Health, School of Public Health, Jilin University, Changchun, China.
| |
Collapse
|
7
|
White UA, Maier J, Zhao P, Richard AJ, Stephens JM. The modulation of adiponectin by STAT5-activating hormones. Am J Physiol Endocrinol Metab 2016; 310:E129-36. [PMID: 26601851 PMCID: PMC4719028 DOI: 10.1152/ajpendo.00068.2015] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 11/23/2015] [Indexed: 11/22/2022]
Abstract
Adiponectin is a hormone secreted from adipocytes that plays an important role in insulin sensitivity and protects against metabolic syndrome. Growth hormone (GH) and prolactin (PRL) are potent STAT5 activators that regulate the expression of several genes in adipocytes. Studies have shown that the secretion of adiponectin from adipose tissue is decreased by treatment with PRL and GH. In this study, we demonstrate that 3T3-L1 adipocytes treated with GH or PRL exhibit a reduction in adiponectin protein levels. Furthermore, we identified three putative STAT5 binding sites in the murine adiponectin promoter and show that only one of these, located at -3,809, binds nuclear protein in a GH- or PRL-dependent manner. Mutation of the STAT5 binding site reduced PRL-dependent protein binding, and supershift analysis revealed that STAT5A and -5B, but not STAT1 and -3, bind to this site in response to PRL. Chromatin immunoprecipitation (IP) analysis demonstrated that only STAT5A, and not STAT1 and -3, bind to the murine adiponectin promoter in a GH-dependent manner in vivo. Adiponectin promoter/reporter constructs were responsive to GH, and chromatin IP analysis reveals that STAT5 binds the adiponectin promoter in vivo following GH stimulation. Overall, these data strongly suggest that STAT5 activators regulate adiponectin transcription through the binding of STAT5 to the -3,809 site that leads to decreased adiponectin expression and secretion. These mechanistic observations are highly consistent with studies in mice and humans that have high GH or PRL levels that are accompanied by lower circulating levels of adiponectin.
Collapse
Affiliation(s)
- Ursula A White
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and
| | - Joel Maier
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Peng Zhao
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| | - Allison J Richard
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and
| | - Jacqueline M Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana; and Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
8
|
Zhao P, Stephens JM. Identification of STAT target genes in adipocytes. JAKSTAT 2014; 2:e23092. [PMID: 24058802 PMCID: PMC3710315 DOI: 10.4161/jkst.23092] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 12/02/2012] [Accepted: 12/03/2012] [Indexed: 02/08/2023] Open
Abstract
Adipocytes play important roles in lipid storage, energy homeostasis and whole body insulin sensitivity. Studies in the last two decades have identified the hormones and cytokines that activate specific STATs in adipocytes in vitro and in vivo. Five of the seven STAT family members are expressed in adipocyte (STATs 1, 3, 5A, 5B and 6). Many transcription factors, including STATs, have been shown to play an important role in adipose tissue development and function. This review will summarize the importance of adipocytes, indicate the cytokines and hormones that utilize the JAK-STAT signaling pathway in fat cells and focus on the identification of STAT target genes in mature adipocytes. To date, specific target genes have been identified for STATs, 1, 5A and 5B, but not for STATs 3 and 6.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Biological Sciences; Louisiana State University; Baton Rouge, LA USA ; Adipocyte Biology Lab; Pennington Biomedical Research Center; Baton Rouge, LA USA
| | | |
Collapse
|
9
|
Xu D, Yin C, Wang S, Xiao Y. JAK-STAT in lipid metabolism of adipocytes. JAKSTAT 2013; 2:e27203. [PMID: 24498541 DOI: 10.4161/jkst.27203] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 11/13/2013] [Accepted: 11/14/2013] [Indexed: 11/19/2022] Open
Abstract
JAK-STAT signaling pathway plays an important role in the cells' development and homeostasis. Over the past decades, the studies have identified the role of the JAK-STAT pathway in cell proliferation and apoptosis. Here, we want to discuss that whether and how the JAK-STAT pathway affects the lipid metabolism of adipose tissue. A host of cytokines and hormones can regulate lipid metabolism through activating the JAK-STAT signaling pathway. Activated STATs can regulate lipid metabolism directly by influencing the expression of enzymes. We have summarized the relevant research and articles of JAK-STAT during the recent years. Within this review, we will introduce you the recent research and highlight the unresolved problems in understanding how JAK-STAT signaling pathway contribute to the lipid metabolism in mature adipocytes and preadipocytes. Dysregulation of the JAK-STAT pathway would lead to a multiple metabolism disorders and medicines for this signaling pathway maybe become a new idea for diseases such as metabolic syndrome, especially in children.
Collapse
Affiliation(s)
- Dong Xu
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| | - Chunyan Yin
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| | - Sisi Wang
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| | - Yanfeng Xiao
- Department of Pediatrics; Second Affiliated Hospital of Medical School of Xi'an; Jiaotong University; Xi'an, Shaanxi PR China
| |
Collapse
|
10
|
The role of JAK-STAT signaling in adipose tissue function. Biochim Biophys Acta Mol Basis Dis 2013; 1842:431-9. [PMID: 23735217 DOI: 10.1016/j.bbadis.2013.05.030] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Revised: 05/20/2013] [Accepted: 05/22/2013] [Indexed: 01/14/2023]
Abstract
Adipocytes play important roles in lipid storage, energy homeostasis and whole body insulin sensitivity. The JAK-STAT (Janus Kinase-Signal Transducer and Activator of Transcription) pathway mediates a variety of physiological processes including development, hematopoiesis, and inflammation. Although the JAK-STAT signaling pathway occurs in all cells, this pathway can mediate cell specific responses. Studies in the last two decades have identified hormones and cytokines that activate the JAK-STAT signaling pathway. These cytokines and hormones have profound effects on adipocytes. The content of this review will introduce the types of adipocytes and immune cells that make up adipose tissue, the impact of obesity on adipose cellular composition and function, and the general constituents of the JAK-STAT pathway and how its activators regulate adipose tissue development and physiology. A summary of the identification of STAT target genes in adipocytes reveals how these transcription factors impact various areas of adipocyte metabolism including insulin action, modulation of lipid stores, and glucose homeostasis. Lastly, we will evaluate exciting new data linking the JAK-STAT pathway and brown adipose tissue and consider the future outlook in this area of investigation. This article is part of a Special Issue entitled: Modulation of Adipose Tissue in Health and Disease.
Collapse
|
11
|
Davoodi-Semiromi A, Wasserfall CH, Hassanzadeh A, Cooper-DeHoff RM, Wabitsch M, Atkinson M. Influence of Tyrphostin AG490 on the expression of diabetes-associated markers in human adipocytes. Immunogenetics 2012; 65:83-90. [PMID: 23081744 DOI: 10.1007/s00251-012-0659-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 10/08/2012] [Indexed: 11/25/2022]
Abstract
Tyrosine kinase inhibitors (TKi) hold promise as a treatment for a variety of disorders ranging from those in oncology to diseases thought as immune mediated. Tyrphostin AG490 is a potent Jak-Stat TKi shown effective in the prevention of allograft transplant rejection, experimental autoimmune disease, as well as the treatment of cancer. However, given its ability to modulate this important but pleiotropic intracellular pathway, we thought that it is important to examine its effects on glucose metabolism and expression of major transcription factors and adipokines associated with insulin insensitivity and diabetes. We investigated the metabolic effects of AG490 on glucose levels in vivo using an animal model of diabetes, nonobese diabetic (NOD) mice, and transcription factor expression through assessment of human adipocytes. AG490 treatment of young nondiabetic NOD mice significantly reduced blood glucose levels (p = 0.002). In vitro, treatment of adipocytes with rosiglitazone, an insulin sensitizer that binds to peroxisome proliferator-activated receptor (PPAR) receptors and increases the adipocyte response to insulin, significantly increased the expression of the antidiabetic adipokine adiponectin. Importantly, the combination of rosiglitazone plus Tyrphostin AG490 further increased this effect and was specifically associated with significant upregulation of C-enhanced binding protein (C/EBP) (p < 0.0001). In terms of the mechanism underlying this action, regulatory regions of the PPARγ, ADIPOQ, and C/EBP contain the Stat5 DNA-binding sequences and were demonstrated, by gel shift experiments in vitro. These data suggest that blocking Jak-Stat signaling with AG490 reduces blood glucose levels and modulates the expression of transcription factors previously associated with diabetes, thereby supporting its potential as a therapy for this disease.
Collapse
Affiliation(s)
- Abdoreza Davoodi-Semiromi
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Gainesville, FL 33136, USA.
| | | | | | | | | | | |
Collapse
|
12
|
Shetty S, Ramos-Roman MA, Cho YR, Brown J, Plutzky J, Muise ES, Horton JD, Scherer PE, Parks EJ. Enhanced fatty acid flux triggered by adiponectin overexpression. Endocrinology 2012; 153:113-22. [PMID: 22045665 PMCID: PMC3249680 DOI: 10.1210/en.2011-1339] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2011] [Accepted: 09/30/2011] [Indexed: 12/30/2022]
Abstract
Adiponectin overexpression in mice increases insulin sensitivity independent of adiposity. Here, we combined stable isotope infusion and in vivo measurements of lipid flux with transcriptomic analysis to characterize fatty acid metabolism in transgenic mice that overexpress adiponectin via the aP2-promoter (ADNTg). Compared with controls, fasted ADNTg mice demonstrated a 31% reduction in plasma free fatty acid concentrations (P = 0.008), a doubling of ketones (P = 0.028), and a 68% increase in free fatty acid turnover in plasma (15.1 ± 1.5 vs. 25.3 ± 6.8 mg/kg · min, P = 0.011). ADNTg mice had 2-fold more brown adipose tissue mass, and triglyceride synthesis and turnover were 5-fold greater in this organ (P = 0.046). Epididymal white adipose tissue was slightly reduced, possibly due to the approximately 1.5-fold increase in the expression of genes involved in oxidation (peroxisome proliferator-activated receptor α, peroxisome proliferator-activated receptor-γ coactivator 1α, and uncoupling protein 3). In ADNTg liver, lipogenic gene expression was reduced, but there was an unexpected increase in the expression of retinoid pathway genes (hepatic retinol binding protein 1 and retinoic acid receptor beta and adipose Cyp26A1) and liver retinyl ester content (64% higher, P < 0.02). Combined, these data support a physiological link between adiponectin signaling and increased efficiency of triglyceride synthesis and hydrolysis, a process that can be controlled by retinoids. Interactions between adiponectin and retinoids may underlie adiponectin's effects on intermediary metabolism.
Collapse
Affiliation(s)
- Shoba Shetty
- Touchstone Diabetes Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390-9052, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Richard AJ, Stephens JM. Emerging roles of JAK-STAT signaling pathways in adipocytes. Trends Endocrinol Metab 2011; 22:325-32. [PMID: 21561789 PMCID: PMC3149764 DOI: 10.1016/j.tem.2011.03.007] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 03/16/2011] [Accepted: 03/30/2011] [Indexed: 01/14/2023]
Abstract
Twenty years ago, adipocytes were largely considered to be inert energy-storage depots. We now know that fat cells are highly insulin-sensitive with significant endocrine functions. Alterations in adipocyte development or function can contribute to metabolic disease, in particular type 2 diabetes. The current obesity epidemic that plagues many nations provides a strong rationale for understanding basic adipocyte biology. The JAK-STAT signaling pathway mediates the action of a variety of hormones that have profound effects on adipocyte development and function. In addition, adipocytes secrete hormones that utilize this signaling pathway. This review summarizes research on the expression and function of JAKs and STATs in adipocytes and highlights the roles of JAK-STAT-activating cytokines in adipose tissue.
Collapse
Affiliation(s)
| | - Jacqueline M. Stephens
- To whom correspondence should be addressed, Louisiana State University, Department of Biological Sciences, 202 Life Sciences Bldg., Baton Rouge, LA 70803, Phone (225)-578-1749, FAX (225)-578-2597,
| |
Collapse
|
14
|
Joseph SJ, Robbins KR, Pavan E, Pratt SL, Duckett SK, Rekaya R. Effect of diet supplementation on the expression of bovine genes associated with Fatty Acid synthesis and metabolism. Bioinform Biol Insights 2010; 4:19-31. [PMID: 20448844 PMCID: PMC2865165 DOI: 10.4137/bbi.s4168] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Conjugated linoleic acids (CLA) are of important nutritional and health benefit to human. Food products of animal origin are their major dietary source and their concentration increases with high concentrate diets fed to animals. To examine the effects of diet supplementation on the expression of genes related to lipid metabolism, 28 Angus steers were fed either pasture only, pasture with soybean hulls and corn oil, pasture with corn grain, or high concentrate diet. At slaughter, samples of subcutaneous adipose tissue were collected, from which RNA was extracted. Relative abundance of gene expression was measured using Affymetrix GeneChip Bovine Genome array. An ANOVA model nested within gene was used to analyze the background adjusted, normalized average difference of probe-level intensities. To control experiment wise error, a false discovery rate of 0.01 was imposed on all contrasts. Expression of several genes involved in the synthesis of enzymes related to fatty acid metabolism and lipogenesis such as stearoyl-CoA desaturase (SCD), fatty acid synthetase (FASN), lipoprotein lipase (LPL), fatty-acyl elongase (LCE) along with several trancription factors and co-activators involved in lipogenesis were found to be differentially expressed. Confirmatory RT-qPCR was done to validate the microarray results, which showed satisfactory correspondence between the two platforms. Results show that changes in diet by increasing dietary energy intake by supplementing high concentrate diet have effects on the transcription of genes encoding enzymes involved in fat metabolism which in turn has effects on fatty acid content in the carcass tissue as well as carcass quality. Corn supplementation either as oil or grain appeared to significantly alter the expression of genes directly associated with fatty acid synthesis.
Collapse
|
15
|
Qian L, Lopez V, Seo YA, Kelleher SL. Prolactin regulates ZNT2 expression through the JAK2/STAT5 signaling pathway in mammary cells. Am J Physiol Cell Physiol 2009; 297:C369-77. [PMID: 19494234 DOI: 10.1152/ajpcell.00589.2008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The zinc transporter ZnT2 (SLC30A2) plays an important role in zinc secretion into milk during lactation. The physiological process of mammary gland secretion is regulated through complex integration of multiple lactogenic hormones. Prolactin plays a primary role in this regulation through the activation of various signaling cascades including Jak2/STAT5, mitogen-activated protein kinase (MAPK), p38, and phosphatidylinositol 3-kinase (PI3K). The precise mechanisms that regulate the transfer of specific nutrients such as zinc into milk are not well understood. Herein we report that prolactin increased ZnT2 abundance transcriptionally in cultured mammary epithelial (HC11) cells. To delineate the responsible mechanisms, we first determined that prolactin-mediated ZnT2 induction was inhibited by pretreatment with the Jak2 inhibitor AG490 but not by the MAPK inhibitor PD-98059. Using a luciferase reporter assay, we demonstrated that ZnT2 promoter activity was increased by prolactin treatment, which was subsequently abolished by expression of a dominant-negative STAT5 construct, implicating the Jak2/STAT5 signaling pathway in the transcriptional regulation of ZnT2. Two putative consensus STAT5 binding sequences in the ZnT2 promoter were identified (GAS1:-674 to -665 and GAS2:-377 to -368). Mutagenesis of the proximal GAS2 element resulted in complete abrogation of PRL-induced ZnT2 promoter activity. The promoter incorporating the distal GAS1 mutation was only able to respond to very high PRL concentrations. Results from both the mutagenesis and gel shift assays indicated that a cooperative relationship exists between GAS1 and GAS2 for PRL-induced activation; however, the proximal GAS2 plays a more critical role in STAT5-mediated signal transduction compared with the GAS1 element. Finally, chromosome immunoprecipition assay further confirmed that prolactin activates STAT5 binding to the ZnT2 promoter in vivo. Taken together, these results illustrate that prolactin regulates the transcription of ZnT2 through activation of the Jak2/STAT5 signaling pathway to assist in providing optimal zinc for secretion into milk during lactation.
Collapse
Affiliation(s)
- Linxi Qian
- Department of Nutritional Sciences, the Pennsylvania State University, University Park, PA 16802, USA
| | | | | | | |
Collapse
|
16
|
Duckett SK, Pratt SL, Pavan E. Corn oil or corn grain supplementation to steers grazing endophyte-free tall fescue. II. Effects on subcutaneous fatty acid content and lipogenic gene expression. J Anim Sci 2008; 87:1120-8. [PMID: 19028850 DOI: 10.2527/jas.2008-1420] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Twenty-eight Angus steers (289 kg) were finished on a high-concentrate diet (85% concentrate: 15% roughage; CONC), or endophyte-free tall fescue pastures with corn grain supplement (0.52% of BW; PC), corn oil plus soybean hull supplement (0.10% of BW corn oil plus 0.45% of BW soybean hulls; PO), or no supplement (pasture only; PA). Subcutaneous adipose tissues were processed for total cellular RNA extraction and fatty acid composition by GLC. Relative expression of genes involved in lipogenesis [fatty acid synthase (FASN), acetyl-CoA carboxylase, lipoprotein lipase, stearoyl-CoA desaturase (SCD)] and activators of transcription [(peroxisome proliferator-activated receptor-gamma), C/EBPalpha, sterol regulatory binding protein-1, signal transducer and activator of transcription-5, and Spot-14] was determined by real-time quantitative PCR. Housekeeping gene (glyceraldehyde 3-phosphate dehydrogenase and beta-actin) expression was used in analysis to normalize expression data. Total fatty acid content was greatest (P < 0.001) for CONC and least (P < 0.001) for PA. Supplementation of grazing cattle increased (P < 0.001) total fatty acid content compared with PA, but concentrations were less (P < 0.001) than for CONC. Myristic and palmitic acid contents were greater (P < 0.001) for CONC than for PO and PC, which were greater (P < 0.001) than for PA. Stearic acid content was greater (P < 0.01) for PO than for CONC, PC, and PA. Finishing on CONC increased (P < 0.001) total MUFA content by 68% compared with PA. Corn grain supplementation increased (P < 0.001) MUFA content compared with PA; in contrast, MUFA content did not differ (P > 0.05) between PO and PA. Corn oil supplementation increased (P < 0.001) trans-11 vaccenic acid content in subcutaneous fat by 1.2-, 1.7- and 5.6-fold relative to PA, PC, and CONC, respectively. Concentrations of the cis-9, trans-11 CLA isomer were 54, 58, and 208% greater (P < 0.01) for PO than for PA, PC, and CONC, respectively. Corn grain supplementation to grazing steers did not alter (P > 0.05) the cis-9, trans-11 CLA isomer compared with PA. Oil supplementation increased (P < 0.001) linoleic acid (C18:2) content by 56, 98 and 262% compared with CONC, PC, and PA, respectively. Relative mRNA expression of SCD was upregulated (P < 0.001) by 46-, 18- and 7-fold, respectively, for CONC, PC, and PO compared with PA. Relative FASN mRNA expression was also upregulated (P = 0.004) by 9- and 5-fold, respectively, for CONC and PC compared with PA. Grain feeding, either on CONC or supplemented on pasture, upregulated FASN and SCD mRNA to increase MUFA and de novo fatty acids in subcutaneous adipose tissue. Upregulation of SCD with grain feeding and reduced tissue CLA concentrations suggest that the decreased CLA concentrations were the result of limited substrate (trans-11 vaccenic acid) availability.
Collapse
Affiliation(s)
- S K Duckett
- Clemson University, Clemson, SC 29634-0311, USA.
| | | | | |
Collapse
|
17
|
White UA, Coulter AA, Miles TK, Stephens JM. The STAT5A-mediated induction of pyruvate dehydrogenase kinase 4 expression by prolactin or growth hormone in adipocytes. Diabetes 2007; 56:1623-9. [PMID: 17360981 DOI: 10.2337/db06-1286] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The purpose of this study was to determine whether pyruvate dehydrogenase kinase (PDK)4 was expressed in adipocytes and whether PDK4 expression was hormonally regulated in fat cells. Both Northern blot and Western blot analyses were conducted on samples isolated from 3T3-L1 adipocytes after various treatments with prolactin (PRL), growth hormone (GH), and/or insulin. Transfection of PDK4 promoter reporter constructs was performed. In addition, glucose uptake measurements were conducted. Our studies demonstrate that PRL and porcine GH can induce the expression of PDK4 in 3T3-L1 adipocytes. Our studies also show that insulin pretreatment can attenuate the ability of these hormones to induce PDK4 mRNA expression. In addition, we identified a hormone-responsive region in the murine PDK4 promoter and characterized a STAT5 binding site in this region that mediates the PRL (sheep) and GH (porcine) induction in PDK4 expression in 3T3-L1 adipocytes. PDK4 is a STAT5A target gene. PRL is a potent inducer of PDK4 protein levels, results in an inhibition of insulin-stimulated glucose transport in fat cells, and likely contributes to PRL-induced insulin resistance.
Collapse
Affiliation(s)
- Ursula A White
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803, USA
| | | | | | | |
Collapse
|