1
|
Liu J, Ma C, Cheng Y, Wang M, Zhao G, Huang L, Song R, Wang X, Li H. METTL14 and WTAP play a crucial role in the regulation of bovine preadipocyte differentiation. Anim Biotechnol 2025; 36:2476531. [PMID: 40094566 DOI: 10.1080/10495398.2025.2476531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 03/19/2025]
Abstract
m6A methylation is the most common mRNA modification in mammals and plays a significant role in regulating various biological functions. Some studies have demonstrated that the methyltransferase METTL3 can promote adipogenesis. However, the regulatory mechanisms of METTL14 and WTAP, both methyltransferases, in adipogenesis remain unclear. This study investigated their effects on bovine preadipocyte differentiation using siRNA-mediated knockdown combined with transcriptomic analysis. Silencing METTL14 and WTAP significantly impaired lipid droplet formation and revealed distinct regulatory pathways: METTL14 knockdown affected genes like JAK2 and STAT3, while WTAP suppression down-regulated PPARγ/FABP4 signalling pathway components. These findings demonstrate that WTAP specifically modulates bovine adipocyte differentiation through the PPARγ/FABP4 pathway.
Collapse
Affiliation(s)
- Jia Liu
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Chicheng Ma
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Yu Cheng
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Minzhi Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Guoqing Zhao
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Liwei Huang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Ruigao Song
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Xi Wang
- College of Animal Science, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Hongxia Li
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
2
|
Wang C, Chen C, Lei B, Qin S, Zhang Y, Li K, Zhang S, Liu Y. Constructing eRNA-mediated gene regulatory networks to explore the genetic basis of muscle and fat-relevant traits in pigs. Genet Sel Evol 2024; 56:28. [PMID: 38594607 PMCID: PMC11003151 DOI: 10.1186/s12711-024-00897-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 04/03/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Enhancer RNAs (eRNAs) play a crucial role in transcriptional regulation. While significant progress has been made in understanding epigenetic regulation mediated by eRNAs, research on the construction of eRNA-mediated gene regulatory networks (eGRN) and the identification of critical network components that influence complex traits is lacking. RESULTS Here, employing the pig as a model, we conducted a comprehensive study using H3K27ac histone ChIP-seq and RNA-seq data to construct eRNA expression profiles from multiple tissues of two distinct pig breeds, namely Enshi Black (ES) and Duroc. In addition to revealing the regulatory landscape of eRNAs at the tissue level, we developed an innovative network construction and refinement method by integrating RNA-seq, ChIP-seq, genome-wide association study (GWAS) signals and enhancer-modulating effects of single nucleotide polymorphisms (SNPs) measured by self-transcribing active regulatory region sequencing (STARR-seq) experiments. Using this approach, we unraveled eGRN that significantly influence the growth and development of muscle and fat tissues, and identified several novel genes that affect adipocyte differentiation in a cell line model. CONCLUSIONS Our work not only provides novel insights into the genetic basis of economic pig traits, but also offers a generalizable approach to elucidate the eRNA-mediated transcriptional regulation underlying a wide spectrum of complex traits for diverse organisms.
Collapse
Affiliation(s)
- Chao Wang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Choulin Chen
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Bowen Lei
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China
| | - Shenghua Qin
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
| | - Yuanyuan Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- School of Life Sciences, Henan University, Kaifeng, 475004, People's Republic of China
- Shenzhen Research Institute of Henan University, Shenzhen, 518000, People's Republic of China
| | - Kui Li
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China
| | - Song Zhang
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China.
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China.
| | - Yuwen Liu
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Key Laboratory of Livestock and Poultry Multi-Omics of MARA, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China.
- Innovation Group of Pig Genome Design and Breeding, Research Centre for Animal Genome, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518124, People's Republic of China.
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction of Ministry of Education & Key Lab of Swine Genetics and Breeding of Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, People's Republic of China.
- Kunpeng Institute of Modern Agriculture at Foshan, Chinese Academy of Agricultural Sciences, Foshan, 528226, People's Republic of China.
| |
Collapse
|
3
|
Perumal NL, Do SK, Choi JS, Lee JH, Ban GT, Kim G, Mufida A, Yoo HS, Jang BC. Anti‑adipogenic effect and underlying mechanism of lignan‑enriched nutmeg extract on 3T3‑L1 preadipocytes. Biomed Rep 2024; 20:4. [PMID: 38124767 PMCID: PMC10729302 DOI: 10.3892/br.2023.1692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/31/2023] [Indexed: 12/23/2023] Open
Abstract
Nutmeg is the seed derived from Myristica fragrans. Nutmeg seeds contain alkylbenzene derivatives such as myristicin, which are toxic to the human organism, and lignan compounds such as nectandrin B, which possess anti-aging and anti-diabetic properties. However, the anti-adipogenic, prolipolytic and anti-inflammatory effects of lignan-enriched nutmeg extract (LNX) on preadipocytes remain unclear. In the present study, the effects of LNX on lipid accumulation, glycerol release and inflammatory cyclooxygenase-2 (COX-2) expression in differentiated 3T3-L1 preadipocytes were investigated. Oil red O staining demonstrated that treatment with LNX resulted in a concentration-dependent reduction in lipid accumulation in differentiating 3T3-L1 preadipocytes without affecting cell growth. Mechanistically, LNX treatment at 6 µg/ml led to a reduction in phosphorylation levels of signal transducer and activator of transcription 3 (STAT3), whereas it did not influence the peroxisome proliferator-activated receptor gamma (PPAR-γ) and CCAAT enhancer binding protein alpha (C/EBP-α) expression levels during 3T3-L1 preadipocyte differentiation. In addition, LNX treatment at 6 µg/ml led to a decrease in fatty acid synthase (FAS) expression levels on day (D) 2, but not D5 and D8, during preadipocyte differentiation. Treatment with LNX at 6 µg/ml did not affect the expression levels of perilipin A during preadipocyte differentiation. In differentiated 3T3-L1 adipocytes, LNX treatment at 6 µg/ml did not stimulate glycerol release and hormone-sensitive lipase phosphorylation, which are known lipolysis hallmarks. Furthermore, LNX treatment at the doses tested had no effect on tumor necrosis factor alpha-induced COX-2 expression in 3T3-L1 preadipocytes. Collectively, these results demonstrated that LNX has an anti-adipogenic effect on differentiating 3T3-L1 preadipocytes, which is mediated by the downregulation of STAT3 phosphorylation and FAS expression.
Collapse
Affiliation(s)
| | - Sung Kuk Do
- College of Korean Medicine, Daejeon University, Daejeon 34520, Republic of Korea
| | - Jong-Soon Choi
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon 34133, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Je-Ho Lee
- Geron Biotech Ltd., Daejeon 34133, Republic of Korea
| | - Gyung-Tae Ban
- Geron Biotech Ltd., Daejeon 34133, Republic of Korea
| | - Gyuri Kim
- Research Center for Materials Analysis, Korea Basic Science Institute, Daejeon 34133, Republic of Korea
- Graduate School of Analytical Science and Technology, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Amila Mufida
- Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| | - Hwa Seung Yoo
- College of Korean Medicine, Daejeon University, Daejeon 34520, Republic of Korea
| | - Byeong-Churl Jang
- Department of Molecular Medicine, College of Medicine, Keimyung University, Daegu 42601, Republic of Korea
| |
Collapse
|
4
|
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
|
5
|
Fetuin-A regulates adipose tissue macrophage content and activation in insulin resistant mice through MCP-1 and iNOS: involvement of IFNγ-JAK2-STAT1 pathway. Biochem J 2021; 478:4027-4043. [PMID: 34724561 DOI: 10.1042/bcj20210442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/29/2021] [Accepted: 11/01/2021] [Indexed: 12/12/2022]
Abstract
In the context of obesity-induced adipose tissue (AT) inflammation, migration of macrophages and their polarization from predominantly anti-inflammatory to proinflammatory subtype is considered a pivotal event in the loss of adipose insulin sensitivity. Two major chemoattractants, monocyte chemoattractant protein-1 (MCP-1) and Fetuin-A (FetA), have been reported to stimulate macrophage migration into inflamed AT instigating inflammation. Moreover, FetA could notably modulate macrophage polarization, yet the mechanism(s) is unknown. The present study was undertaken to elucidate the mechanistic pathway involved in the actions of FetA and MCP-1 in obese AT. We found that FetA knockdown in high fat diet (HFD) fed mice could significantly subdue the augmented MCP-1 expression and reduce adipose tissue macrophage (ATM) content thereby indicating that MCP-1 is being regulated by FetA. Additionally, knockdown of FetA in HFD mice impeded the expression of inducible nitric oxide synthase (iNOS) reverting macrophage activation from mostly proinflammatory to anti-inflammatory state. It was observed that the stimulating effect of FetA on MCP-1 and iNOS was mediated through interferon γ (IFNγ) induced activation of JAK2-STAT1-NOX4 pathway. Furthermore, we detected that the enhanced IFNγ expression was accounted by the stimulatory effect of FetA upon the activities of both cJun and JNK. Taken together, our findings revealed that obesity-induced FetA acts as a master upstream regulator of AT inflammation by regulating MCP-1 and iNOS expression through JNK-cJun-IFNγ-JAK2-STAT1 signaling pathway. This study opened a new horizon in understanding the regulation of ATM content and activation in conditions of obesity-induced insulin resistance.
Collapse
|
6
|
Koh E, Kim B, Choi K. Torreya nucifera seed oil improves 3T3-L1 adipocyte differentiation. BMC Complement Med Ther 2021; 21:255. [PMID: 34620154 PMCID: PMC8496151 DOI: 10.1186/s12906-021-03429-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 09/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Adipose tissue is a critical regulator of lipid storage and endocrine function. Impairment of the recruitment of new adipocytes in the adipose tissue is associated with ectopic fat accumulation, diabetes and insulin resistance. Torreya nucifera, an evergreen conifer that grows in warm temperate climates, has been found to exert beneficial effects against inflammation, infection and diabetes. However, the molecular mechanisms responsible for these effects at the cellular level remain unknown. This study aimed to investigate effects of Torreya nucifera seed oil (TNSO) on 3T3-L1 adipocyte differentiation and its underlying regulatory mechanism. METHODS To investigate the effects of TNSO on adipocyte differentiation, 3T3-L1 cells were induced to differentiate for 5 days in the presence of 0.75 μL/mL TNSO. Oil Red O staining and an assay for intracellular triglyceride were performed to determine the extent of lipid accumulation in 3T3-L1 cells. To elucidate the underlying mechanism of TNSO, adipogenic gene expression was analyzed using quantitative real-time PCR. Moreover, we monitored TNSO-derived activation of PPARγ and STAT3 with 3T3-L1 reporter cell lines engineered to secrete Gaussia luciferase upon the interaction of a transcription factor to its DNA binding element. RESULTS Oil Red O staining revealed that TNSO improved the differentiation of 3T3-L1 preadipocytes into mature adipocytes. The mRNA levels of adipogenic genes, including adiponectin, fatty acid synthase (FAS) and adipocyte fatty acid-binding protein (FABP4), were upregulated and intracellular triglyceride levels increased upon TNSO treatment. We also established that adipocyte differentiation was improved by TNSO-derived activation of PPARγ and STAT3. CONCLUSIONS Our results suggest that TNSO improves adipocyte differentiation by regulating the activation of adipogenic transcription factors, indicating that it may serve as a potential treatment strategy for adipocyte dysfunction.
Collapse
Affiliation(s)
- Eunbi Koh
- Department of Chemical and Material Engineering, The University of Suwon, 17, Wauan-gil, Bongdam-eup, Hwaseong-si, Gyeonggi-do, 18323, Republic of Korea
| | - Boram Kim
- Department of Chemical and Material Engineering, The University of Suwon, 17, Wauan-gil, Bongdam-eup, Hwaseong-si, Gyeonggi-do, 18323, Republic of Korea
| | - Kyungoh Choi
- Department of Chemical and Material Engineering, The University of Suwon, 17, Wauan-gil, Bongdam-eup, Hwaseong-si, Gyeonggi-do, 18323, Republic of Korea.
| |
Collapse
|
7
|
Lipocalin-2: Structure, function, distribution and role in metabolic disorders. Biomed Pharmacother 2021; 142:112002. [PMID: 34463264 DOI: 10.1016/j.biopha.2021.112002] [Citation(s) in RCA: 187] [Impact Index Per Article: 46.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/26/2021] [Accepted: 08/01/2021] [Indexed: 12/27/2022] Open
Abstract
Lipocalin-2 (LCN-2) is a novel, 198 amino acid adipocytokine also referred to as neutrophil gelatinase-associated lipocalin (NGAL). LCN-2 is a circulatory protein responsible for the transportation of small and hydrophobic molecules (steroid, free fatty acids, prostaglandins and hormones) to target organs after binding to megalin/glycoprotein and GP330 SLC22A17 or 24p3R LCN-2 receptors. LCN-2 has been used as a biomarker for acute and chronic renal injury. It is present in a large variety of cells including neutrophil, hepatocytes, lung, bone marrow, adipose tissue, macrophages, thymus, non-neoplastic breast duct, prostate, and renal cells. Different functions have been associated with LCN-2. These functions include antibacterial, anti-inflammatory, and protection against cell and tissue stress. Moreover, LCN-2 can increase the pool of matrix metalloproteinase 9 in human neutrophil granulocytes. Other reported functions of LCN-2 include its ability to destroy the extracellular matrix, which could enable cancer progression and spread of metastasis. Recent reports show that the tissue level of LCN-2 is increased in metabolic disorders such as obesity and type 2 diabetes, suggesting an association between LCN-2 and insulin sensitivity and glucose homeostasis. The precise role of LCN-2 in the modulation of insulin sensitivity, glucose and lipid metabolism is still unclear. This review explores the structure of LCN-2, tissue distribution, and its interaction with important metabolic pathways.
Collapse
|
8
|
Alves CDS, Frias HV, Bonamin LV, Correia MSF, Corrêa MG, Bondan EF, de Fátima M Martins M, Coelho CP, Bernardi MM, Suffredini IB. Luffa operculata at a late period of gestation dysregulates melatonin and cytokines interfering with weight of dams and their male offspring. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:113867. [PMID: 33892067 DOI: 10.1016/j.jep.2021.113867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/20/2020] [Accepted: 01/18/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The tea made with the fruits of Luffa operculata (L.) Cogn. (Cucurbitaceae; EBN) is popularly used as abortive. AIM OF THE STUDY The present work aimed at accessing how the exposition of female Wistar rats to 1.0 mg/kg of EBN (experimental group, EG), or distilled water (control group, CG), by gavage, at gestational days (GD) 17-21 interfered with the reproductive performance, and with dams' behavior after weaning. MATERIALS AND METHODS At post-natal day 2 (PND2), the number of male and female pups was evaluated, as well as their weight. After weaning (PND21), dams were euthanized, and their liver and kidneys were removed for histological and biochemical analyses, while the blood was used in the evaluation of cytokines IL-1α, IL-1β, IL-6 and TNF-α, corticosterone, adrenocorticotrophic hormone, melatonin, AST, ALT and creatinine levels. RESULTS AND DISCUSSION Dams that were treated with EBN showed an anxiety-like behavior, weight loss at the end of gestation and weight gain at weaning, accompanied with a significant decrease in pro-inflammatory cytokines and in the melatonin level. No significant histological or biochemical alterations have occurred in the liver or kidneys. The number of female pups was significantly higher in the EG. The male pups showed weight gain at PND60. CONCLUSION The presence of cucurbitacins is probably involved in the dysregulations that were found, due to their polycyclic steroid triterpene structure.
Collapse
Affiliation(s)
- Cinthia Dos S Alves
- Programa de Pós-Graduação Em Patologia Ambiental e Experimental, Universidade Paulista - UNIP, Brazil
| | - Humberto V Frias
- Programa de Pós-Graduação Em Patologia Ambiental e Experimental, Universidade Paulista - UNIP, Brazil
| | - Leoni V Bonamin
- Programa de Pós-Graduação Em Patologia Ambiental e Experimental, Universidade Paulista - UNIP, Brazil
| | - Michelle S F Correia
- Programa de Pós-Graduação Em Patologia Ambiental e Experimental, Universidade Paulista - UNIP, Brazil
| | - Monica G Corrêa
- Programa de Pós-Graduação Em Odontologia, Universidade Paulista - UNIP, Brazil
| | - Eduardo F Bondan
- Programa de Pós-Graduação Em Patologia Ambiental e Experimental, Universidade Paulista - UNIP, Brazil
| | | | | | - Maria M Bernardi
- Programa de Pós-Graduação Em Patologia Ambiental e Experimental, Universidade Paulista - UNIP, Brazil; Programa de Pós-Graduação Em Odontologia, Universidade Paulista - UNIP, Brazil
| | - Ivana B Suffredini
- Programa de Pós-Graduação Em Patologia Ambiental e Experimental, Universidade Paulista - UNIP, Brazil; Programa de Pós-Graduação Em Odontologia, Universidade Paulista - UNIP, Brazil; Núcleo de Pesquisas Em Biodiversidade, Laboratório de Extração, Universidade Paulista - UNIP, Brazil.
| |
Collapse
|
9
|
Qin A, Chen S, Wang P, Huang X, Zhang Y, Liang L, Du LR, Lai DH, Ding L, Yu X, Xiang AP. Knockout of NOS2 Promotes Adipogenic Differentiation of Rat MSCs by Enhancing Activation of JAK/STAT3 Signaling. Front Cell Dev Biol 2021; 9:638518. [PMID: 33816486 PMCID: PMC8017136 DOI: 10.3389/fcell.2021.638518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/08/2021] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) are a heterogeneous population of cells that possess multilineage differentiation potential and extensive immunomodulatory properties. In mice and rats, MSCs produce nitric oxide (NO), as immunomodulatory effector molecule that exerts an antiproliferative effect on T cells, while the role of NO in differentiation was less clear. Here, we investigated the role of NO synthase 2 (NOS2) on adipogenic and osteogenic differentiation of rat MSCs. MSCs isolated from NOS2-null (NOS2–/–) and wild type (WT) Sprague–Dawley (SD) rats exhibited homogenous fibroblast-like morphology and characteristic phenotypes. However, after induction, adipogenic differentiation was found significantly promoted in NOS2–/– MSCs compared to WT MSCs, but not in osteogenic differentiation. Accordingly, qRT-PCR revealed that the adipogenesis-related genes PPAR-γ, C/EBP-α, LPL and FABP4 were markedly upregulated in NOS2–/– MSCs, but not for osteogenic transcription factors or marker genes. Further investigations revealed that the significant enhancement of adipogenic differentiation in NOS2–/– MSCs was due to overactivation of the STAT3 signaling pathway. Both AG490 and S3I-201, small molecule inhibitors that selectively inhibit STAT3 activation, reversed this adipogenic effect. Furthermore, after high-fat diet (HFD) feeding, knockout of NOS2 in rat MSCs resulted in significant obesity. In summary, NOS2 is involved in the regulation of rat MSC adipogenic differentiation via the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Aiping Qin
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China.,Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Sheng Chen
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ping Wang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Xiaotao Huang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Yu Zhang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Lu Liang
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Ling-Ran Du
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - De-Hua Lai
- Center for Parasitic Organisms, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Li Ding
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiyong Yu
- Key Laboratory of Molecular Target and Clinical Pharmacology, State Key Laboratory of Respiratory Disease, The Fifth Affiliated Hospital, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
10
|
Zhao P, Saltiel AR. Interaction of Adipocyte Metabolic and Immune Functions Through TBK1. Front Immunol 2020; 11:592949. [PMID: 33193441 PMCID: PMC7606291 DOI: 10.3389/fimmu.2020.592949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Adipocytes and adipose tissue play critical roles in the regulation of metabolic homeostasis. In obesity and obesity-associated metabolic diseases, immune cells infiltrate into adipose tissues. Interaction between adipocytes and immune cells re-shapes both metabolic and immune properties of adipose tissue and dramatically changes metabolic set points. Both the expression and activity of the non-canonical IKK family member TBK1 are induced in adipose tissues during diet-induced obesity. TBK1 plays important roles in the regulation of both metabolism and inflammation in adipose tissue and thus affects glucose and energy metabolism. Here we review the regulation and functions of TBK1 and the molecular mechanisms by which TBK1 regulates both metabolism and inflammation in adipose tissue. Finally, we discuss the potential of a TBK1/IKKε inhibitor as a new therapy for metabolic diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Alan R Saltiel
- Department of Medicine, University of California San Diego, La Jolla, CA, United States.,Department of Pharmacology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
11
|
Latest advances in STAT signaling and function in adipocytes. Clin Sci (Lond) 2020; 134:629-639. [PMID: 32219346 DOI: 10.1042/cs20190522] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/24/2020] [Accepted: 03/09/2020] [Indexed: 02/07/2023]
Abstract
Adipocytes and adipose tissue are not inert and make substantial contributions to systemic metabolism by influencing energy homeostasis, insulin sensitivity, and lipid storage. In addition to well-studied hormones such as insulin, there are numerous hormones, cytokines, and growth factors that modulate adipose tissue function. Many endocrine mediators utilize the JAK-STAT pathway to mediate dozens of biological processes, including inflammation and immune responses. JAKs and STATs can modulate both adipocyte development and mature adipocyte function. Of the seven STAT family members, four STATs are expressed in adipocytes and regulated during adipogenesis (STATs 1, 3, 5A, and 5B). These STATs have been shown to play influential roles in adipose tissue development and function. STAT6, in contrast, is highly expressed in both preadipocytes and mature adipocytes, but is not considered to play a major role in regulating adipose tissue function. This review will summarize the latest research that pertains to the functions of STATs in adipocytes and adipose tissue.
Collapse
|
12
|
Steuernagel L, Meckbach C, Heinrich F, Zeidler S, Schmitt AO, Gültas M. Computational identification of tissue-specific transcription factor cooperation in ten cattle tissues. PLoS One 2019; 14:e0216475. [PMID: 31095599 PMCID: PMC6522001 DOI: 10.1371/journal.pone.0216475] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 04/22/2019] [Indexed: 01/01/2023] Open
Abstract
Transcription factors (TFs) are a special class of DNA-binding proteins that orchestrate gene transcription by recruiting other TFs, co-activators or co-repressors. Their combinatorial interplay in higher organisms maintains homeostasis and governs cell identity by finely controlling and regulating tissue-specific gene expression. Despite the rich literature on the importance of cooperative TFs for deciphering the mechanisms of individual regulatory programs that control tissue specificity in several organisms such as human, mouse, or Drosophila melanogaster, to date, there is still need for a comprehensive study to detect specific TF cooperations in regulatory processes of cattle tissues. To address the needs of knowledge about specific combinatorial gene regulation in cattle tissues, we made use of three publicly available RNA-seq datasets and obtained tissue-specific gene (TSG) sets for ten tissues (heart, lung, liver, kidney, duodenum, muscle tissue, adipose tissue, colon, spleen and testis). By analyzing these TSG-sets, tissue-specific TF cooperations of each tissue have been identified. The results reveal that similar to the combinatorial regulatory events of model organisms, TFs change their partners depending on their biological functions in different tissues. Particularly with regard to preferential partner choice of the transcription factors STAT3 and NR2C2, this phenomenon has been highlighted with their five different specific cooperation partners in multiple tissues. The information about cooperative TFs could be promising: i) to understand the molecular mechanisms of regulating processes; and ii) to extend the existing knowledge on the importance of single TFs in cattle tissues.
Collapse
Affiliation(s)
- Lukas Steuernagel
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany
| | - Cornelia Meckbach
- Institute of Medical Bioinformatics, Goldschmidtstraße 1, University Medical Center Göttingen, Georg-August-University, 37077 Göttingen, Germany
| | - Felix Heinrich
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany
| | - Sebastian Zeidler
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany
| | - Armin O. Schmitt
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany
- Center for Integrated Breeding Research (CiBreed), Albrecht-Thaer-Weg 3, Georg-August University, 37075, Göttingen, Germany
| | - Mehmet Gültas
- Breeding Informatics Group, Department of Animal Sciences, Georg-August University, Margarethe von Wrangell-Weg 7, 37075 Göttingen, Germany
- Center for Integrated Breeding Research (CiBreed), Albrecht-Thaer-Weg 3, Georg-August University, 37075, Göttingen, Germany
- * E-mail:
| |
Collapse
|
13
|
Allum F, Hedman ÅK, Shao X, Cheung WA, Vijay J, Guénard F, Kwan T, Simon MM, Ge B, Moura C, Boulier E, Rönnblom L, Bernatsky S, Lathrop M, McCarthy MI, Deloukas P, Tchernof A, Pastinen T, Vohl MC, Grundberg E. Dissecting features of epigenetic variants underlying cardiometabolic risk using full-resolution epigenome profiling in regulatory elements. Nat Commun 2019; 10:1209. [PMID: 30872577 PMCID: PMC6418220 DOI: 10.1038/s41467-019-09184-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 02/25/2019] [Indexed: 12/16/2022] Open
Abstract
Sparse profiling of CpG methylation in blood by microarrays has identified epigenetic links to common diseases. Here we apply methylC-capture sequencing (MCC-Seq) in a clinical population of ~200 adipose tissue and matched blood samples (Ntotal~400), providing high-resolution methylation profiling (>1.3 M CpGs) at regulatory elements. We link methylation to cardiometabolic risk through associations to circulating plasma lipid levels and identify lipid-associated CpGs with unique localization patterns in regulatory elements. We show distinct features of tissue-specific versus tissue-independent lipid-linked regulatory regions by contrasting with parallel assessments in ~800 independent adipose tissue and blood samples from the general population. We follow-up on adipose-specific regulatory regions under (1) genetic and (2) epigenetic (environmental) regulation via integrational studies. Overall, the comprehensive sequencing of regulatory element methylomes reveals a rich landscape of functional variants linked genetically as well as epigenetically to plasma lipid traits.
Collapse
Affiliation(s)
- Fiona Allum
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Åsa K Hedman
- Department of Medicine Solna, Cardiovascular Medicine Unit, Karolinska Institute, Stockholm, 171 76, Sweden
| | - Xiaojian Shao
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Warren A Cheung
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
- Children's Mercy Hospitals and Clinics, Kansas City, MO, 64108, USA
| | - Jinchu Vijay
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Frédéric Guénard
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC, G1V 0A6, Canada
| | - Tony Kwan
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Marie-Michelle Simon
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Bing Ge
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Cristiano Moura
- Department of Epidemiology, McGill University, Montréal, QC, H3A 1A2, Canada
| | - Elodie Boulier
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Lars Rönnblom
- Department of Medical Sciences, Uppsala University, Uppsala, 751 85, Sweden
| | - Sasha Bernatsky
- Department of Epidemiology, McGill University, Montréal, QC, H3A 1A2, Canada
| | - Mark Lathrop
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
| | - Mark I McCarthy
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, University of Oxford, Old Road, Headington, Oxford, OX3 7LJ, UK
- Wellcome Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford, OX3 7BN, UK
- Oxford NIHR Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK
| | - André Tchernof
- Québec Heart and Lung Institute, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Tomi Pastinen
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada
- Children's Mercy Hospitals and Clinics, Kansas City, MO, 64108, USA
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods (INAF), Université Laval, Québec, QC, G1V 0A6, Canada
| | - Elin Grundberg
- Department of Human Genetics, McGill University, Montréal, QC, H3A 0C7, Canada.
- McGill University and Genome Quebec Innovation Centre, Montréal, QC, H3A 0G1, Canada.
- Children's Mercy Hospitals and Clinics, Kansas City, MO, 64108, USA.
| |
Collapse
|
14
|
Murtaza M, Khan G, Aftab MF, Afridi SK, Ghaffar S, Ahmed A, Hafizur RM, Waraich RS. Cucurbitacin E reduces obesity and related metabolic dysfunction in mice by targeting JAK-STAT5 signaling pathway. PLoS One 2017; 12:e0178910. [PMID: 28598969 PMCID: PMC5466318 DOI: 10.1371/journal.pone.0178910] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/19/2017] [Indexed: 01/14/2023] Open
Abstract
Several members of cucurbitaceae family have been reported to regulate growth of cancer by interfering with STAT3 signaling. In the present study, we investigated the unique role and molecular mechanism of cucurbitacins (Cucs) in reducing symptoms of metabolic syndrome in mice. Cucurbitacin E (CuE) was found to reduce adipogenesis in murine adipocytes. CuE treatment diminished hypertrophy of adipocytes, visceral obesity and lipogenesis gene expression in diet induced mice model of metabolic syndrome (MetS). CuE also ameliorated adipose tissue dysfunction by reducing hyperleptinemia and TNF-alpha levels and enhancing hypoadiponectinemia. Results show that CuE mediated these effects by attenuating Jenus kinase- Signal transducer and activator of transcription 5 (JAK- STAT5) signaling in visceral fat tissue. As a result, CuE treatment also reduced PPAR gamma expression. Glucose uptake enhanced in adipocytes after stimulation with CuE and insulin resistance diminished in mice treated with CuE, as reflected by reduced glucose intolerance and glucose stimulated insulin secretion. CuE restored insulin sensitivity indirectly by inhibiting JAK phosphorylation and improving AMPK activity. Consequently, insulin signaling was up-regulated in mice muscle. As CuE positively regulated adipose tissue function and suppressed visceral obesity, dyslipedemia, hyperglycemia and insulin resistance in mice model of MetS, we suggest that CuE can be used as novel approach to treat metabolic diseases.
Collapse
Affiliation(s)
- Munazza Murtaza
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Gulnaz Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Meha Fatima Aftab
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Shabbir Khan Afridi
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Safina Ghaffar
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Ayaz Ahmed
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rahman M Hafizur
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| | - Rizwana Sanaullah Waraich
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan
| |
Collapse
|
15
|
Ambele MA, Pepper MS. Identification of transcription factors potentially involved in human adipogenesis in vitro. Mol Genet Genomic Med 2017; 5:210-222. [PMID: 28546992 PMCID: PMC5441431 DOI: 10.1002/mgg3.269] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/26/2016] [Accepted: 12/09/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Increased adiposity in humans leads to obesity, which is a major risk factor for cardiovascular disease, type 2 diabetes, and cancer. We previously conducted an extensive unbiased in vitro transcriptomic analysis of adipogenesis, using human adipose-derived stromal cells (ASCs). Here, we have applied computational methods to these data to identify transcription factors (TFs) that constitute the upstream gene regulatory networks potentially, driving adipocyte formation in human ASCs. METHODS We used Affymetrix Transcription Analysis Console™ v3.0 for calculating differentially expressed genes. MATCH™ and F-MATCH™ algorithms for TF identification. STRING v10 to predict protein-protein interactions between TFs. RESULTS A number of TFs that were reported to have a significant role in adipogenesis, as well as novel TFs that have not previously been described in this context, were identified. Thus, 32 upstream TFs were identified, with most belonging to the C2H2-type zinc finger and HOX families, which are potentially involved in regulating most of the differentially expressed genes observed during adipocyte differentiation. Furthermore, 17 important upstream TFs were found to have increased regulatory effects on their downstream target genes and were consistently up-regulated during the differentiation process. A strong hypothetical functional interaction was observed among these TFs, which supports their common role in the downstream regulation of gene expression during adipogenesis. CONCLUSION Our results support several previous findings on TFs involved in adipogenesis and thereby validate the comprehensive and systematic in silico approach described in this study. In silico analysis also allowed for the identification of novel regulators of adipocyte differentiation.
Collapse
Affiliation(s)
- Melvin Anyasi Ambele
- Department of Immunology and Institute for Cellular and Molecular MedicineSAMRC Extramural Unit for Stem Cell Research and TherapyFaculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Michael Sean Pepper
- Department of Immunology and Institute for Cellular and Molecular MedicineSAMRC Extramural Unit for Stem Cell Research and TherapyFaculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| |
Collapse
|
16
|
Kramer AH, Kadye R, Houseman PS, Prinsloo E. Mitochondrial STAT3 and reactive oxygen species: A fulcrum of adipogenesis? JAKSTAT 2015; 4:e1084084. [PMID: 27127727 DOI: 10.1080/21623996.2015.1084084] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/07/2015] [Accepted: 08/11/2015] [Indexed: 02/08/2023] Open
Abstract
The balance between cellular lineages can be controlled by reactive oxygen species (ROS). Cellular differentiation into adipocytes is highly dependent on the production of ROS to initiate the process through activation of multiple interlinked factors that stimulate mitotic clonal expansion and cellular maturation. The signal transducer and activator of transcription family of signaling proteins have accepted roles in adipogenesis and associated lipogenesis. Non-canonical mitochondrial localization of STAT3 and other members of the STAT family however opens up new avenues for investigation of its role in the aforementioned processes. Following recent observations of differences in mitochondrially localized serine 727 phosphorylated STAT3 (mtSTAT3-pS727) in preadipocytes and adipocytes, here, we hypothesize and speculate further on the role of mitochondrial STAT3 in adipogenesis.
Collapse
Affiliation(s)
- Adam H Kramer
- Biotechnology Innovation Center; Rhodes University ; Grahamstown, South Africa
| | - Rose Kadye
- Biotechnology Innovation Center; Rhodes University ; Grahamstown, South Africa
| | | | - Earl Prinsloo
- Biotechnology Innovation Center; Rhodes University ; Grahamstown, South Africa
| |
Collapse
|
17
|
Zhao P, Elks CM, Stephens JM. The induction of lipocalin-2 protein expression in vivo and in vitro. J Biol Chem 2014; 289:5960-9. [PMID: 24391115 DOI: 10.1074/jbc.m113.532234] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Lipocalin-2 (LCN2) is secreted from adipocytes, and its expression is up-regulated in obese and diabetic mice and humans. LCN2 expression and secretion have been shown to be induced by two proinflammatory cytokines, IFNγ and TNFα, in cultured murine and human adipocytes. In these studies, we demonstrated that IFNγ and TNFα induced LCN2 expression and secretion in vivo. Although we observed a strong induction of LCN2 expression and secretion from white adipose tissue (WAT) depots, the induction of LCN2 varied among different insulin-sensitive tissues. Knockdown experiments also demonstrated that STAT1 is required for IFNγ-induced lipocalin-2 expression in murine adipocytes. Similarly, knockdown of p65 in adipocytes demonstrated the necessity of the NF-κB signaling pathway for TNFα-mediated effects on LCN2. Activation of ERKs by IFNγ and TNFα also affected STAT1 and NF-κB signaling through modulation of serine phosphorylation. ERK activation-induced serine phosphorylation of both STAT1 and p65 mediated the additive effects of IFNγ and TNFα on LCN2 expression. Our results suggest that these same mechanisms occur in humans as we observed STAT1 and NF-κB binding to the human LCN2 promoter in chromatin immunoprecipitation assays performed in human fat cells. These studies substantially increase our knowledge regarding the requirements and mechanisms used by proinflammatory cytokines to induce LCN2 expression.
Collapse
Affiliation(s)
- Peng Zhao
- From the Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana 70803 and Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808
| | | | | |
Collapse
|
18
|
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
|