1
|
Guo G, Wang W, Tu M, Zhao B, Han J, Li J, Pan Y, Zhou J, Ma W, Liu Y, Sun T, Han X, An Y. Deciphering adipose development: Function, differentiation and regulation. Dev Dyn 2024; 253:956-997. [PMID: 38516819 DOI: 10.1002/dvdy.708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/23/2024] Open
Abstract
The overdevelopment of adipose tissues, accompanied by excess lipid accumulation and energy storage, leads to adipose deposition and obesity. With the increasing incidence of obesity in recent years, obesity is becoming a major risk factor for human health, causing various relevant diseases (including hypertension, diabetes, osteoarthritis and cancers). Therefore, it is of significance to antagonize obesity to reduce the risk of obesity-related diseases. Excess lipid accumulation in adipose tissues is mediated by adipocyte hypertrophy (expansion of pre-existing adipocytes) or hyperplasia (increase of newly-formed adipocytes). It is necessary to prevent excessive accumulation of adipose tissues by controlling adipose development. Adipogenesis is exquisitely regulated by many factors in vivo and in vitro, including hormones, cytokines, gender and dietary components. The present review has concluded a comprehensive understanding of adipose development including its origin, classification, distribution, function, differentiation and molecular mechanisms underlying adipogenesis, which may provide potential therapeutic strategies for harnessing obesity without impairing adipose tissue function.
Collapse
Affiliation(s)
- Ge Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wanli Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Mengjie Tu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Binbin Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiayang Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jiali Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yanbing Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Wen Ma
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yi Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Tiantian Sun
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Xu Han
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| | - Yang An
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- Henan Provincial Engineering Center for Tumor Molecular Medicine, Kaifeng Key Laboratory of Cell Signal Transduction, Henan University, Kaifeng, China
| |
Collapse
|
2
|
Liao Y, Zhang W, Liu Y, Zhu C, Zou Z. The role of ubiquitination in health and disease. MedComm (Beijing) 2024; 5:e736. [PMID: 39329019 PMCID: PMC11424685 DOI: 10.1002/mco2.736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/28/2024] Open
Abstract
Ubiquitination is an enzymatic process characterized by the covalent attachment of ubiquitin to target proteins, thereby modulating their degradation, transportation, and signal transduction. By precisely regulating protein quality and quantity, ubiquitination is essential for maintaining protein homeostasis, DNA repair, cell cycle regulation, and immune responses. Nevertheless, the diversity of ubiquitin enzymes and their extensive involvement in numerous biological processes contribute to the complexity and variety of diseases resulting from their dysregulation. The ubiquitination process relies on a sophisticated enzymatic system, ubiquitin domains, and ubiquitin receptors, which collectively impart versatility to the ubiquitination pathway. The widespread presence of ubiquitin highlights its potential to induce pathological conditions. Ubiquitinated proteins are predominantly degraded through the proteasomal system, which also plays a key role in regulating protein localization and transport, as well as involvement in inflammatory pathways. This review systematically delineates the roles of ubiquitination in maintaining protein homeostasis, DNA repair, genomic stability, cell cycle regulation, cellular proliferation, and immune and inflammatory responses. Furthermore, the mechanisms by which ubiquitination is implicated in various pathologies, alongside current modulators of ubiquitination are discussed. Enhancing our comprehension of ubiquitination aims to provide novel insights into diseases involving ubiquitination and to propose innovative therapeutic strategies for clinical conditions.
Collapse
Affiliation(s)
- Yan Liao
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Wangzheqi Zhang
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Yang Liu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Chenglong Zhu
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| | - Zui Zou
- Faculty of Anesthesiology Changhai Hospital Naval Medical University Shanghai China
- School of Anesthesiology Naval Medical University Shanghai China
| |
Collapse
|
3
|
Chao X, Guo L, Ye C, Liu A, Wang X, Ye M, Fan Z, Luan K, Chen J, Zhang C, Liu M, Zhou B, Zhang X, Li Z, Luo Q. ALKBH5 regulates chicken adipogenesis by mediating LCAT mRNA stability depending on m 6A modification. BMC Genomics 2024; 25:634. [PMID: 38918701 PMCID: PMC11197345 DOI: 10.1186/s12864-024-10537-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND Previous studies have demonstrated the role of N6-methyladenosine (m6A) RNA methylation in various biological processes, our research is the first to elucidate its specific impact on LCAT mRNA stability and adipogenesis in poultry. RESULTS The 6 100-day-old female chickens were categorized into high (n = 3) and low-fat chickens (n = 3) based on their abdominal fat ratios, and their abdominal fat tissues were processed for MeRIP-seq and RNA-seq. An integrated analysis of MeRIP-seq and RNA-seq omics data revealed 16 differentially expressed genes associated with to differential m6A modifications. Among them, ELOVL fatty acid elongase 2 (ELOVL2), pyruvate dehydrogenase kinase 4 (PDK4), fatty acid binding protein 9 (PMP2), fatty acid binding protein 1 (FABP1), lysosomal associated membrane protein 3 (LAMP3), lecithin-cholesterol acyltransferase (LCAT) and solute carrier family 2 member 1 (SLC2A1) have ever been reported to be associated with adipogenesis. Interestingly, LCAT was down-regulated and expressed along with decreased levels of mRNA methylation methylation in the low-fat group. Mechanistically, the highly expressed ALKBH5 gene regulates LCAT RNA demethylation and affects LCAT mRNA stability. In addition, LCAT inhibits preadipocyte proliferation and promotes preadipocyte differentiation, and plays a key role in adipogenesis. CONCLUSIONS In conclusion, ALKBH5 mediates RNA stability of LCAT through demethylation and affects chicken adipogenesis. This study provides a theoretical basis for further understanding of RNA methylation regulation in chicken adipogenesis.
Collapse
Affiliation(s)
- Xiaohuan Chao
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lijin Guo
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Chutian Ye
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Aijun Liu
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xiaomeng Wang
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mao Ye
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhexia Fan
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Kang Luan
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiahao Chen
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Chunlei Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Manqing Liu
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Bo Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiquan Zhang
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China
- College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Zhenhui Li
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China.
- College of Animal Science, South China Agricultural University, Guangzhou, China.
| | - Qingbin Luo
- State Key Laboratory of Livestock and Poultry Breeding, South China Agricultural University, Guangzhou, China.
- College of Animal Science, South China Agricultural University, Guangzhou, China.
| |
Collapse
|
4
|
Xiao Y, Xie X, Chen Z, Yin G, Kong W, Zhou J. Advances in the roles of ATF4 in osteoporosis. Biomed Pharmacother 2023; 169:115864. [PMID: 37948991 DOI: 10.1016/j.biopha.2023.115864] [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: 09/07/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023] Open
Abstract
Osteoporosis (OP) is characterized by reduced bone mass, decreased strength, and enhanced bone fragility fracture risk. Activating transcription factor 4 (ATF4) plays a role in cell differentiation, proliferation, apoptosis, redox balance, amino acid uptake, and glycolipid metabolism. ATF4 induces the differentiation of bone marrow mesenchymal stem cells (BM-MSCs) into osteoblasts, increases osteoblast activity, and inhibits osteoclast formation, promoting bone formation and remodeling. In addition, ATF4 mediates the energy metabolism in osteoblasts and promotes angiogenesis. ATF4 is also involved in the mediation of adipogenesis. ATF4 can selectively accumulate in osteoblasts. ATF4 can directly interact with RUNT-related transcription factor 2 (RUNX2) and up-regulate the expression of osteocalcin (OCN) and osterix (Osx). Several upstream factors, such as Wnt/β-catenin and BMP2/Smad signaling pathways, have been involved in ATF4-mediated osteoblast differentiation. ATF4 promotes osteoclastogenesis by mediating the receptor activator of nuclear factor κ-B (NF-κB) ligand (RANKL) signaling. Several agents, such as parathyroid (PTH), melatonin, and natural compounds, have been reported to regulate ATF4 expression and mediate bone metabolism. In this review, we comprehensively discuss the biological activities of ATF4 in maintaining bone homeostasis and inhibiting OP development. ATF4 has become a therapeutic target for OP treatment.
Collapse
Affiliation(s)
- Yaosheng Xiao
- Department of Orthopaetics, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xunlu Xie
- Department of Pathology, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Zhixi Chen
- Department of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Guoqiang Yin
- Ganzhou Hospital Affiliated to Nanchang University, Ganzhou 341000, China
| | - Weihao Kong
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China
| | - Jianguo Zhou
- Department of Joint Surgery, Ganzhou People's Hospital, Ganzhou 341000, China.
| |
Collapse
|
5
|
Song Y, Wei D, Raza SHA, Zhao Y, Jiang C, Song X, Wu H, Wang X, Luoreng Z, Ma Y. Research progress of intramuscular fat formation based on co-culture. Anim Biotechnol 2023; 34:3216-3236. [PMID: 36200856 DOI: 10.1080/10495398.2022.2127410] [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] [Indexed: 11/01/2022]
Abstract
Intramuscular fat (IMF) is closely related to the meat quality of livestock and poultry. As a new cell culture technique in vitro, cell co-culture has been gradually applied to the related research of IMF formation because it can simulate the changes of microenvironment in vivo during the process of IMF cell formation. In the co-culture model, in addition to studying the effects of skeletal muscle cells on the proliferation and differentiation of IMF, we can also consider the role of many secretion factors in the formation of IMF, thus making the cell research in vitro closer to the real level in vivo. This paper reviewed the generation and origin of IMF, summarized the existing co-culture methods and systems, and discussed the advantages and disadvantages of each method as well as the challenges faced in the establishment of the system, with emphasis on the current status of research on the formation of IMF for human and animal based on co-culture technology.
Collapse
Affiliation(s)
- Yaping Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Dawei Wei
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | | | - Yiang Zhao
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Chao Jiang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xiaoyu Song
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Hao Wu
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Xingping Wang
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Zhuoma Luoreng
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Ningxia Yin Chuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia University, Ningxia Yinchuan, China
| |
Collapse
|
6
|
Kim Y, Kim HK, Kang S, Kim H, Go GW. Rottlerin suppresses lipid accumulation by inhibiting de novo lipogenesis and adipogenesis via LRP6/mTOR/SREBP1C in 3T3-L1 adipocytes. Food Sci Biotechnol 2023; 32:1445-1452. [PMID: 37457404 PMCID: PMC10349001 DOI: 10.1007/s10068-023-01339-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/24/2023] [Accepted: 05/12/2023] [Indexed: 07/18/2023] Open
Abstract
Rottlerin is isolated from Mallotus japonicus, a plant rich in polyphenols. Rottlerin is a selective PKCδ-inhibitor and is also known as an uncoupler of oxidative phosphorylation and anti-neoplastic agent. However, its anti-obesity effect is yet to be established. Therefore, this study tested whether rottlerin inhibits adipogenesis and de novo lipogenesis via the LRP6/mTOR/SREBP1C pathway in 3T3-L1 adipocytes. Rottlerin dramatically decreased lipid accumulation assessed by Oil Red O as evidence to support the cellular phenotype (p < 0.001). Pivotal messenger RNA and protein expressions associated with de novo lipogenesis (SREBP1C, ACC1, FAS, and SCD1) and adipogenesis (PPARγ and C/EBPα) were subsequentially verified by rottlerin in a dose-dependent manner (p < 0.05). Further investigation revealed that rottlerin reduced the AKT/mTOR pathway via diminished total protein of LRP6 (p < 0.05). Collectively, these findings establish a causal link between rottlerin, LRP6, and the altered nutrient-sensing mTOR pathway, in which rottlerin regulates de novo lipogenesis and adipogenesis in white adipocytes.
Collapse
Affiliation(s)
- Yejin Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Hyun Kyung Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Sumin Kang
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Hayoon Kim
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| | - Gwang-woong Go
- Department of Food and Nutrition, Hanyang University, Seoul, 04763 Republic of Korea
| |
Collapse
|
7
|
Aloke C, Iwuchukwu EA, Achilonu I. Exploiting Copaifera salikounda compounds as treatment against diabetes: An insight into their potential targets from a computational perspective. Comput Biol Chem 2023; 104:107851. [DOI: 10.1016/j.compbiolchem.2023.107851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/25/2023] [Accepted: 03/19/2023] [Indexed: 03/29/2023]
|
8
|
Hansen GT, Sobreira DR, Weber ZT, Thornburg AG, Aneas I, Zhang L, Sakabe NJ, Joslin AC, Haddad GA, Strobel SM, Laber S, Sultana F, Sahebdel F, Khan K, Li YI, Claussnitzer M, Ye L, Battaglino RA, Nóbrega MA. Genetics of sexually dimorphic adipose distribution in humans. Nat Genet 2023; 55:461-470. [PMID: 36797366 PMCID: PMC10375400 DOI: 10.1038/s41588-023-01306-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 01/23/2023] [Indexed: 02/18/2023]
Abstract
Obesity-associated morbidity is exacerbated by abdominal obesity, which can be measured as the waist-to-hip ratio adjusted for the body mass index (WHRadjBMI). Here we identify genes associated with obesity and WHRadjBMI and characterize allele-sensitive enhancers that are predicted to regulate WHRadjBMI genes in women. We found that several waist-to-hip ratio-associated variants map within primate-specific Alu retrotransposons harboring a DNA motif associated with adipocyte differentiation. This suggests that a genetic component of adipose distribution in humans may involve co-option of retrotransposons as adipose enhancers. We evaluated the role of the strongest female WHRadjBMI-associated gene, SNX10, in adipose biology. We determined that it is required for human adipocyte differentiation and function and participates in diet-induced adipose expansion in female mice, but not males. Our data identify genes and regulatory mechanisms that underlie female-specific adipose distribution and mediate metabolic dysfunction in women.
Collapse
Affiliation(s)
- Grace T Hansen
- Department of Human Genetics, University of Chicago, Chicago, IL, USA.
- Pritzker School of Medicine, University of Chicago, Chicago, IL, USA.
| | - Débora R Sobreira
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Zachary T Weber
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | | | - Ivy Aneas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Li Zhang
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Noboru J Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Amelia C Joslin
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Gabriela A Haddad
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Sophie M Strobel
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Institute of Nutritional Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Samantha Laber
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford, UK
| | - Farhath Sultana
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Faezeh Sahebdel
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Kohinoor Khan
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA
| | - Yang I Li
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
- Department of Genetic Medicine, University of Chicago, Chicago, IL, USA
| | - Melina Claussnitzer
- Broad Institute of MIT and Harvard, Boston, MA, USA
- Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA, USA
- Massachussetts General Hospital, Harvard Medical School, Boston, MA, USA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease at the Broad Institute of MIT and Harvard, Boston, MA, USA
| | - Liang Ye
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA.
| | - Ricardo A Battaglino
- Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, USA.
| | - Marcelo A Nóbrega
- Department of Human Genetics, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
9
|
Kitamura H. Ubiquitin-Specific Proteases (USPs) and Metabolic Disorders. Int J Mol Sci 2023; 24:3219. [PMID: 36834633 PMCID: PMC9966627 DOI: 10.3390/ijms24043219] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 02/10/2023] Open
Abstract
Ubiquitination and deubiquitination are reversible processes that modify the characteristics of target proteins, including stability, intracellular localization, and enzymatic activity. Ubiquitin-specific proteases (USPs) constitute the largest deubiquitinating enzyme family. To date, accumulating evidence indicates that several USPs positively and negatively affect metabolic diseases. USP22 in pancreatic β-cells, USP2 in adipose tissue macrophages, USP9X, 20, and 33 in myocytes, USP4, 7, 10, and 18 in hepatocytes, and USP2 in hypothalamus improve hyperglycemia, whereas USP19 in adipocytes, USP21 in myocytes, and USP2, 14, and 20 in hepatocytes promote hyperglycemia. In contrast, USP1, 5, 9X, 14, 15, 22, 36, and 48 modulate the progression of diabetic nephropathy, neuropathy, and/or retinopathy. USP4, 10, and 18 in hepatocytes ameliorates non-alcoholic fatty liver disease (NAFLD), while hepatic USP2, 11, 14, 19, and 20 exacerbate it. The roles of USP7 and 22 in hepatic disorders are controversial. USP9X, 14, 17, and 20 in vascular cells are postulated to be determinants of atherosclerosis. Moreover, mutations in the Usp8 and Usp48 loci in pituitary tumors cause Cushing syndrome. This review summarizes the current knowledge about the modulatory roles of USPs in energy metabolic disorders.
Collapse
Affiliation(s)
- Hiroshi Kitamura
- Laboratory of Comparative Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu 069-8501, Japan
| |
Collapse
|
10
|
Differential Expression Analysis of tRNA-Derived Small RNAs from Subcutaneous Adipose Tissue of Obese and Lean Pigs. Animals (Basel) 2022; 12:ani12243561. [PMID: 36552481 PMCID: PMC9774726 DOI: 10.3390/ani12243561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/12/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
Epigenetic factors, including non-coding RNA regulation, play a vital role in the development of obesity and have been well researched. Transfer RNA-derived small RNA (tsRNA) is a class of non-coding RNA proven to be involved in various aspects of mammalian biology. Here we take pigs as a model for obesity research and use tsRNA-seq to investigate the difference in tsRNA expression in the subcutaneous adipose tissue of obese and lean pigs to elucidate the role of tsRNA in obesity development. A total of 482 tsRNAs were identified in pig adipose tissue, of which 123 were significantly differentially accumulated tsRNAs compared with the control group. The tRF-5c was the main type of these tsRNAs. The largest number of tsRNAs produced was the Gly-carrying tRNA, which produced 81 tsRNAs. Functional enrichment analysis revealed that differential tsRNAs indirectly participated in MAPK, AMPK, insulin resistance, the TNF signaling pathway, adipocytokine signaling pathway, and other signaling pathways by interacting with target genes. These are involved in bioenergetic metabolic regulatory processes, suggesting that tsRNAs may influence these pathways to mediate the regulation of energy metabolism in porcine adipocytes to promote lipid deposition, thus contributing to obesity. Our findings suggest a potential function of tsRNA in regulating obesity development.
Collapse
|
11
|
Identification and Validation of Ferroptosis-Related Genes in Sevoflurane-Induced Hippocampal Neurotoxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4435161. [PMID: 36238640 PMCID: PMC9553355 DOI: 10.1155/2022/4435161] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/25/2022] [Indexed: 11/17/2022]
Abstract
Background Sevoflurane is one of the most popular inhalational anesthetics during perioperative period but presenting neurotoxicity among pediatric and aged populations. Recent experiments in vivo and in vitro have indicated that ferroptosis may contribute to the neurotoxicity of sevoflurane anesthesia. However, the exact mechanism is still unclear. Methods In current study, we explored the differential expressed genes (DEGs) in HT-22 mouse hippocampal neuronal cells after sevoflurane anesthesia using RNA-seq. Differential expressed ferroptosis-related genes (DEFRGs) were screened and analyzed by Gene Ontology (GO) and pathway enrichment analysis. Protein-to-protein interaction (PPI) network was constructed by the Search Tool for the Retrieval of Interacting Genes (STRING). Significant modules and the hub genes were identified by using Cytoscape. The Connectivity Map (cMAP) was used for screening drug candidates targeting the identified DEFRGs. Potential TF-gene network and drug-gene pairs were established towards the hub genes. In final, we validated these results in experiments. Results A total of 37 ferroptosis-related genes (18 upregulated and 19 downregulated) after sevoflurane exposure in hippocampal neuronal cells were finally identified. These differentially expressed genes were mainly involved into the biological processes of cellular response to oxidative stress. Pathway analysis indicated that these genes were involved in ferroptosis, mTOR signaling pathway, and longevity-regulating pathway. PPI network was constructed. 10 hub genes including Prkaa2, Chac1, Arntl, Tfrc, Slc7a11, Atf4, Mgst1, Lpin1, Atf3, and Sesn2 were found. Top 10 drug candidates, gene-drug networks, and TFs targeting these genes were finally identified. These results were validated in experiments. Conclusion Our results suggested that ferroptosis-related genes play roles in sevoflurane anesthesia-related hippocampal neuron injury and offered the hub genes and potential therapeutic agents for investigating and treatment of this neurotoxicity after sevoflurane exposure. Finally, therapeutic effect of these drug candidates and function of potential ferroptosis targets should be further investigated for treatment and clarifying mechanisms of sevoflurane anesthesia-induced neuron injury in future research.
Collapse
|
12
|
Choi M, Mukherjee S, Yun JW. Colchicine stimulates browning via antagonism of GABA receptor B and agonism of β3-adrenergic receptor in 3T3-L1 white adipocytes. Mol Cell Endocrinol 2022; 552:111677. [PMID: 35598717 DOI: 10.1016/j.mce.2022.111677] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/20/2022]
Abstract
Colchicine has been used for therapeutic purposes and has attracted considerable attention because of its association with tubulin and the inhibition of small tubular polymerization. Although several studies have examined the possible preventive role of colchicine in metabolic diseases, its role in adipocytes is largely unknown. This study examined the novel functional role of colchicine in adipocytes demonstrating that colchicine stimulates browning in cultured white adipocytes. Colchicine stimulates browning by increasing the brown- and beige fat-specific markers in 3T3-L1 white adipocytes. Interestingly, colchicine decreased the expression of the main lipolytic proteins (ATGL, p-HSL) while it activated Ces3, suggesting a possibility for supplying essential fatty acids for inducing thermogenesis. Molecular docking analysis showed that colchicine has a strong affinity against GABA-BR and β3-AR, and its binding activity with GABA-BR (-26.52 kJ/mol) was stronger than β3-AR (-20.71 kJ/mol). Mechanistic studies were conducted by treating the cells separately with agonists and antagonists of GABA-BR and β3-AR to understand the molecular mechanism underlying the browning effect of colchicine. The results showed that colchicine stimulates browning via the antagonism of GABA-BR and the agonism of β3-AR in 3T3-L1 white adipocytes. The colchicine-mediated activation of β3-AR stimulated the PKA/p38 MAPK signaling pathway, where consequently ATF2 acted as a positive regulator, but AFT4 was a negative regulator for the induction of browning.
Collapse
Affiliation(s)
- MinJi Choi
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea
| | - Sulagna Mukherjee
- Laboratory of Metabolic Signaling,Institute of Bioengineering, School of Life Sciences, EPFL, CH-1015 Lausanne, Switzerland.
| | - Jong Won Yun
- Department of Biotechnology, Daegu University, Gyeongsan, Gyeongbuk, 38453, Republic of Korea.
| |
Collapse
|
13
|
Downregulation of HULC Induces Ferroptosis in Hepatocellular Carcinoma via Targeting of the miR-3200-5p/ATF4 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9613095. [PMID: 35615577 PMCID: PMC9126659 DOI: 10.1155/2022/9613095] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 04/12/2022] [Accepted: 04/30/2022] [Indexed: 12/20/2022]
Abstract
Hepatocellular carcinoma is a malignant tumor that poses a serious threat to human health. Ferroptosis, which represents an identified type of regulated iron-dependent cell death, may play an important role in hepatocellular carcinoma. However, it is unclear as to whether ferroptosis is involved with the mechanisms of lncRNA HULC in liver cancer cells. Here, we show that knockdown of HULC increases ferroptosis and oxidative stress in liver cancer cells. We also found changes in some related miRNAs in cells treated with HULC siRNA. Differential miRNA expression levels were determined with the use of high-throughput sequencing and prediction target genes identified using bioinformatics analysis. HULC was found to function as a ceRNA of miR-3200-5p, and miR-3200-5p regulates ferroptosis by targeting ATF4, resulting in the inhibition of proliferation and metastasis within HCC cells. In summary, these findings illuminate some of the molecular mechanisms through which downregulation of HULC induces liver cancer cell ferroptosis by targeting the miR-3200-5p/ATF4 axis to modulate the development of hepatocellular carcinoma.
Collapse
|
14
|
Ku HC, Chan TY, Chung JF, Kao YH, Cheng CF. The ATF3 inducer protects against diet-induced obesity via suppressing adipocyte adipogenesis and promoting lipolysis and browning. Biomed Pharmacother 2022; 145:112440. [PMID: 34839254 DOI: 10.1016/j.biopha.2021.112440] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/10/2021] [Accepted: 11/12/2021] [Indexed: 12/22/2022] Open
Abstract
In this study, we investigated whether the activating transcription factor 3 (ATF3) inducer ST32db, a synthetic compound with a chemical structure similar to that of native Danshen compounds, exerts an anti-obesity effect in 3T3-L1 white preadipocytes, D16 beige cells, and mice with obesity induced by a high-fat diet (HFD). The results showed that ST32db inhibited 3T3-L1 preadipocyte differentiation by inhibiting adipogenesis/lipogenesis-related gene (and protein levels) and enhancing lipolysis-related gene (and protein levels) via the activation of β3-adrenoceptor (β3-AR)/PKA/p38, AMPK, and ERK pathways. Furthermore, ST32db inhibited triacylglycerol accumulation in D16 adipocytes by suppressing adipogenesis/lipogenesis-related gene (and protein levels) and upregulating browning gene expression by suppressing the β3-AR/PKA/p38, and AMPK pathways. Intraperitoneally injected ST32db (1 mg kg-1 twice weekly) inhibited body weight gain and reduced the weight of inguinal white adipose tissue (iWAT), epididymal WAT (eWAT), and mesenteric WAT, with no effects on food intake by the obese mice. The adipocyte diameter and area of iWAT and eWAT were decreased in obese mice injected with ST32db compared with those administered only HFD. In addition, ST32db significantly suppressed adipogenesis and activated lipolysis, browning, mitochondrial oxidative phosphorylation, and β-oxidation-related pathways by suppressing the p38 pathway in the iWAT of the obese mice. These results indicated that the ATF3 inducer ST32db has therapeutic potential for reducing obesity.
Collapse
Affiliation(s)
- Hui-Chen Ku
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
| | - Tsai-Yun Chan
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
| | - Jia-Fang Chung
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan
| | - Yung-Hsi Kao
- Department of Life Sciences, National Central University, Taoyuan 320, Taiwan
| | - Ching-Feng Cheng
- Department of Pediatrics, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City 23142, Taiwan; Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan; Department of Pediatrics, Tzu Chi University, Hualien 97004, Taiwan.
| |
Collapse
|
15
|
Campos JTADM, Oliveira MSD, Soares LP, Medeiros KAD, Campos LRDS, Lima JG. DNA repair-related genes and adipogenesis: Lessons from congenital lipodystrophies. Genet Mol Biol 2022; 45:e20220086. [DOI: 10.1590/1678-4685-gmb-2022-0086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 09/20/2022] [Indexed: 11/09/2022] Open
|
16
|
Lin T, Zhang Y, Zhang T, Steckler RA, Yang X. Hop2 interacts with the transcription factor CEBPα and suppresses adipocyte differentiation. J Biol Chem 2021; 297:101264. [PMID: 34600885 PMCID: PMC8528721 DOI: 10.1016/j.jbc.2021.101264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 09/09/2021] [Accepted: 09/28/2021] [Indexed: 11/16/2022] Open
Abstract
CCAAT enhancer binding protein (CEBP) transcription factors (TFs) are known to promote adipocyte differentiation; however, suppressors of CEBP TFs have not been reported thus far. Here, we find that homologous chromosome pairing protein 2 (Hop2) functions as an inhibitor for the TF CEBPα. We found that Hop2 mRNA is highly and specifically expressed in adipose tissue, and that ectopic Hop2 expression suppresses reporter activity induced by CEBP as revealed by DNA transfection. Recombinant and ectopically expressed Hop2 was shown to interact with CEBPα in pull-down and coimmunoprecipitation assays, and interaction between endogenous Hop2 and CEBPα was observed in the nuclei of 3T3 preadipocytes and adipocytes by immunofluorescence and coimmunoprecipitation of nuclear extracts. In addition, Hop2 stable overexpression in 3T3 preadipocytes inhibited adipocyte differentiation and adipocyte marker gene expression. These in vitro data suggest that Hop2 inhibits adipogenesis by suppressing CEBP-mediated transactivation. Consistent with a negative role for Hop2 in adipogenesis, ablation of Hop2 (Hop2-/-) in mice led to increased body weight, adipose volume, adipocyte size, and adipogenic marker gene expression. Adipogenic differentiation of isolated adipose-derived mesenchymal stem cells showed a greater number of lipid droplet-containing colonies formed in Hop2-/- adipose-derived mesenchymal stem cell cultures than in wt controls, which is associated with the increased expression of adipogenic marker genes. Finally, chromatin immunoprecipitation revealed a higher binding activity of endogenous CEBPα to peroxisome proliferator-activated receptor γ, a master adipogenic TF, and a known CEBPα target gene. Therefore, our study identifies for the first time that Hop2 is an intrinsic suppressor of CEBPα and thus adipogenesis in adipocytes.
Collapse
Affiliation(s)
- Tonghui Lin
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Yang Zhang
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Tingting Zhang
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Rita A Steckler
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA
| | - Xiangli Yang
- Department of Pediatrics, Pediatric Research Center, University of Texas Health Science Center at Houston, McGovern Medical School, Houston, Texas, USA.
| |
Collapse
|
17
|
Hao L, Zhong W, Dong H, Guo W, Sun X, Zhang W, Yue R, Li T, Griffiths A, Ahmadi AR, Sun Z, Song Z, Zhou Z. ATF4 activation promotes hepatic mitochondrial dysfunction by repressing NRF1-TFAM signalling in alcoholic steatohepatitis. Gut 2021; 70:1933-1945. [PMID: 33177163 PMCID: PMC8110597 DOI: 10.1136/gutjnl-2020-321548] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Mitochondrial dysfunction plays a dominant role in the pathogenesis of alcoholic liver disease (ALD); however, the underlying mechanisms remain to be fully understood. We previously found that hepatic activating transcription factor 4 (ATF4) activation was associated with mitochondrial dysfunction in ALD. This study aimed to investigate the function and mechanism of ATF4 in alcohol-induced hepatic mitochondrial dysfunction. DESIGN ATF4 activation was detected in the livers of patients with severe alcoholic hepatitis (AH). The role of ATF4 and mitochondrial transcription factor A (TFAM) in alcohol-induced liver damage was determined in hepatocyte-specific ATF4 knockout mice and liver-specific TFAM overexpression mice, respectively. RESULTS Hepatic PERK-eIF2α-ATF4 ER stress signalling was upregulated in patients with AH. Hepatocyte-specific ablation of ATF4 in mice ameliorated alcohol-induced steatohepatitis. ATF4 ablation also attenuated alcohol-impaired mitochondrial biogenesis and respiratory function along with the restoration of TFAM. Cell studies confirmed that TFAM expression was negatively regulated by ATF4. TFAM silencing in hepatoma cells abrogated the protective effects of ATF4 knockdown on ethanol-mediated mitochondrial dysfunction and cell death. Moreover, hepatocyte-specific TFAM overexpression in mice attenuated alcohol-induced mitochondrial dysfunction and liver damage. Mechanistic studies revealed that ATF4 repressed the transcription activity of nuclear respiratory factor 1 (NRF1), a key regulator of TFAM, through binding to its promoter region. Clinical relevance among ATF4 activation, NRF1-TFAM pathway disruption and mitochondrial dysfunction was validated in the livers of patients with AH. CONCLUSION This study demonstrates that hepatic ATF4 plays a pathological role in alcohol-induced mitochondrial dysfunction and liver injury by disrupting the NRF1-TFAM pathway.
Collapse
Affiliation(s)
- Liuyi Hao
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
| | - Wei Zhong
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
- Department of Nutrition, UNCG, Greensboro, North Carolina, USA
| | - Haibo Dong
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
| | - Wei Guo
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
| | - Xinguo Sun
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
| | - Wenliang Zhang
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
| | - Ruichao Yue
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
| | - Tianjiao Li
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
| | | | - Ali Reza Ahmadi
- Department of Surgery, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Zhaoli Sun
- Department of Surgery, Johns Hopkins Medicine, Baltimore, Maryland, USA
| | - Zhenyuan Song
- Department of Kinesiology and Nutrition, UIC, Chicago, Illinois, USA
| | - Zhanxiang Zhou
- Center for Translational Biomedical Research, UNCG, Kannapolis, North Carolina, USA
- Department of Nutrition, UNCG, Greensboro, North Carolina, USA
| |
Collapse
|
18
|
Chen Y, He R, Han Z, Wu Y, Wang Q, Zhu X, Huang Z, Ye J, Tang Y, Huang H, Chen J, Shan H, Xiao F. Cooperation of ATF4 and CTCF promotes adipogenesis through transcriptional regulation. Cell Biol Toxicol 2021; 38:741-763. [PMID: 33950334 DOI: 10.1007/s10565-021-09608-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
Adipogenesis is a multi-step process orchestrated by activation of numerous TFs, whose cooperation and regulatory network remain elusive. Activating transcription factor 4 (ATF4) is critical for adipogenesis, yet its regulatory network is unclarified. Here, we mapped genome-wide ATF4 binding landscape and its regulatory network by Chip-seq and RNA-seq and found ATF4 directly modulated transcription of genes enriching in fat cell differentiation. Motifs of TFs especially CTCF were found from ATF4 binding sites, suggesting a direct role of ATF4 in regulating adipogenesis associated with CTCF and other TFs. Deletion of CTCF attenuated adipogenesis while overexpression enhanced adipocyte differentiation, indicating CTCF is indispensable for adipogenesis. Intriguingly, combined analysis of Chip-seq data of these two TFs showed that ATF4 co-localized with CTCF in the promoters of key adipogenic genes including Cebpd and PPARg and co-regulated their transactivation. Moreover, ATF4 directly regulated CTCF expression and interacted with CTCF in differentiated 3T3-L1 cells. In vivo, downregulation of ATF4 suppressed the expression of CTCF, Cebpd, and PPARg, leading to reduced adipose tissue expansion in refeeding mice. Consistently, mRNA expression of ATF4 and CTCF was positively correlated with each other in human subcutaneous adipose tissue and inversely associated with BMI, indicating a possible involvement of these two TFs in adipose development. Taken together, our data propose for the first time that ATF4 and CTCF work cooperatively to control adipogenesis and adipose development via orchestrating transcription of adipogenic genes. Our findings reveal novel therapeutic targets in obesity treatment.
Collapse
Affiliation(s)
- Yingchun Chen
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China.,Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Rongquan He
- Department of Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Zhiqiang Han
- Department of Plastic and Aesthetic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, People's Republic of China
| | - Yanyan Wu
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Qiuyan Wang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Xiujuan Zhu
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Zhiguang Huang
- Center for Genomic and Personalized Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 53002, People's Republic of China
| | - Juan Ye
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Yao Tang
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Hongbin Huang
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Jianxu Chen
- Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China
| | - Hong Shan
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China.
| | - Fei Xiao
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China. .,Department of Infectious Diseases, the Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, 519000, People's Republic of China.
| |
Collapse
|
19
|
J U C, Mohan MC, Prakash Kumar B. Attenuation of obesity related inflammation in RAW 264.7 macrophages and 3T3-L1 adipocytes by varanadi kashayam and identification of potential bioactive molecules by UHPLC-Q-Orbitrap HRMS. Arch Physiol Biochem 2021:1-15. [PMID: 33539199 DOI: 10.1080/13813455.2021.1877309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Obesity is associated with chronic low-grade inflammation, characterised by the up-regulation of pro-inflammatory cytokines in obese adipose tissue. In this study, 3T3-L1 CM enhanced TNF-α and IL-1β in RAW 264.7 cells compared with LPS treated cells. However, treatment with Varanadi kashayam suppressed the inflammatory changes associated with RAW 264.7 cells. Subsequently, RAW CM used to stimulate adipocytes, resulting in decreased intracellular lipid content and reduced adipogenic markers after Varanadi kashayam treatment. The chemical profiling of Varanadi kashayam using UHPLC-Q-Orbitrap-HRMS identified 194 compounds by comparing their retention time, the experimentally measured exact mass of precursor, and fragmented ions, and fragmentation pattern with spectral library and reported literature. Collectively, Varanadi kashayam act as a potent anti-inflammatory and anti-adipogenic agent that could disrupt the crosstalk between adipocytes and macrophages. Hence it could be a better candidate for reducing inflammation associated with obese adipose tissue.
Collapse
Affiliation(s)
- Chinchu J U
- Inflammation Research Lab, School of Biosciences, Mahatma Gandhi University, Kottayam, India
| | - Mohind C Mohan
- Inflammation Research Lab, School of Biosciences, Mahatma Gandhi University, Kottayam, India
| | - B Prakash Kumar
- Inflammation Research Lab, School of Biosciences, Mahatma Gandhi University, Kottayam, India
| |
Collapse
|
20
|
Liu H, Li B, Qiao L, Liu J, Ren D, Liu W. miR-340-5p inhibits sheep adipocyte differentiation by targeting ATF7. Anim Sci J 2020; 91:e13462. [PMID: 33190272 DOI: 10.1111/asj.13462] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 08/02/2020] [Accepted: 08/13/2020] [Indexed: 01/01/2023]
Abstract
Several microRNAs (miRNAs) have been identified to play roles in adipocyte differentiation. However, little is known about their involvement in the differentiation of ovine intramuscular adipocytes. Here, the role of one such miRNA, miR-340-5p, in ovine adipocyte differentiation was investigated. Stromal vascular (SV) cells were isolated from skeletal muscle tissues of 1-month-old lambs and induced to differentiate into mature adipocytes. miRNA mimics and inhibitors were used for miR-340-5p overexpression and knockdown assays. For overexpression and knockdown of activating transcription factor 7 (ATF7), lentivirus infection was performed. Luciferase reporter assay was performed to determine the relationship between miR-340-5p and ATF7. The expression of adipogenesis marker genes, PPARγ, C/EBPα, FABP4, ADIPOQ, and ACC, and formation of lipid droplets were detected after the overexpression and inhibition of miR-340-5p, or upon overexpression or knockdown of ATF7. miR-340-5p inhibited the expression of the marker genes and the formation of lipid droplets. ATF7 positively regulated the expression of the marker genes and the formation of lipids. Thus, ATF7 is the target of miR-340-5p in sheep. Overall, these findings indicate that miR-340-5p acts as an inhibitor of the differentiation of intramuscular adipocytes by targeting ATF7. Our study provides a new theoretical basis for improving sheep meat quality.
Collapse
Affiliation(s)
- Haodong Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Baojun Li
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Liying Qiao
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Jianhua Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Duanyang Ren
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| | - Wenzhong Liu
- College of Animal Science and Veterinary Medicine, Shanxi Agricultural University, Taigu, China
| |
Collapse
|
21
|
Criado-Mesas L, Ballester M, Crespo-Piazuelo D, Castelló A, Fernández AI, Folch JM. Identification of eQTLs associated with lipid metabolism in Longissimus dorsi muscle of pigs with different genetic backgrounds. Sci Rep 2020; 10:9845. [PMID: 32555447 PMCID: PMC7300017 DOI: 10.1038/s41598-020-67015-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Accepted: 05/29/2020] [Indexed: 12/14/2022] Open
Abstract
Intramuscular fat content and its fatty acid composition affect porcine meat quality and its nutritional value. The present work aimed to identify genomic variants regulating the expression in the porcine muscle (Longissimus dorsi) of 45 candidate genes for lipid metabolism and fatty acid composition in three experimental backcrosses based on the Iberian breed. Expression genome-wide association studies (eGWAS) were performed between the muscle gene expression values, measured by real-time quantitative PCR, and the genotypes of 38,426 SNPs distributed along all chromosomes. The eGWAS identified 186 eSNPs located in ten Sus scrofa regions and associated with the expression of ACSM5, ACSS2, ATF3, DGAT2, FOS and IGF2 (FDR < 0.05) genes. Two expression quantitative trait loci (eQTLs) for IGF2 and ACSM5 were classified as cis-acting eQTLs, suggesting a mutation in the same gene affecting its expression. Conversely, ten eQTLs showed trans-regulatory effects on gene expression. When the eGWAS was performed for each backcross independently, only three common trans-eQTL regions were observed, indicating different regulatory mechanisms or allelic frequencies among the breeds. In addition, hotspot regions regulating the expression of several genes were detected. Our results provide new data to better understand the functional regulatory mechanisms of lipid metabolism genes in muscle.
Collapse
Affiliation(s)
- Lourdes Criado-Mesas
- Departament de Genòmica Animal, Centre de Recerca en Agrigenòmica (CRAG), CSIC-IRTA-UAB-UB, Barcelona, Spain.
| | - Maria Ballester
- Departament de Genètica i Millora Animal, Institut de Recerca y Tecnologia Agraroalimentàries (IRTA), Caldes de Montbui, Spain
| | - Daniel Crespo-Piazuelo
- Departament de Genòmica Animal, Centre de Recerca en Agrigenòmica (CRAG), CSIC-IRTA-UAB-UB, Barcelona, Spain
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, UAB, Bellaterra, Spain
| | - Anna Castelló
- Departament de Genòmica Animal, Centre de Recerca en Agrigenòmica (CRAG), CSIC-IRTA-UAB-UB, Barcelona, Spain
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, UAB, Bellaterra, Spain
| | - Ana I Fernández
- Departamento de Mejora Genética Animal, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria (INIA), Madrid, Spain
| | - Josep M Folch
- Departament de Genòmica Animal, Centre de Recerca en Agrigenòmica (CRAG), CSIC-IRTA-UAB-UB, Barcelona, Spain
- Departament de Ciència Animal i dels Aliments, Facultat de Veterinària, UAB, Bellaterra, Spain
| |
Collapse
|
22
|
Matsumoto N, Tanaka S, Horiike T, Shinmyo Y, Kawasaki H. A discrete subtype of neural progenitor crucial for cortical folding in the gyrencephalic mammalian brain. eLife 2020; 9:54873. [PMID: 32312384 PMCID: PMC7173966 DOI: 10.7554/elife.54873] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 04/01/2020] [Indexed: 12/28/2022] Open
Abstract
An increase in the diversity of neural progenitor subtypes and folding of the cerebral cortex are characteristic features which appeared during the evolution of the mammalian brain. Here, we show that the expansion of a specific subtype of neural progenitor is crucial for cortical folding. We found that outer radial glial (oRG) cells can be subdivided by HOPX expression in the gyrencephalic cerebral cortex of ferrets. Compared with HOPX-negative oRG cells, HOPX-positive oRG cells had high self-renewal activity and were accumulated in prospective gyral regions. Using our in vivo genetic manipulation technique for ferrets, we found that the number of HOPX-positive oRG cells and their self-renewal activity were regulated by sonic hedgehog (Shh) signaling. Importantly, suppressing Shh signaling reduced HOPX-positive oRG cells and cortical folding, while enhancing it had opposing effects. Our results reveal a novel subtype of neural progenitor important for cortical folding in gyrencephalic mammalian cerebral cortex.
Collapse
Affiliation(s)
- Naoyuki Matsumoto
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Satoshi Tanaka
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.,Medical Research Training Program, School of Medicine, Kanazawa University, Kanazawa, Japan
| | - Toshihide Horiike
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yohei Shinmyo
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Kawasaki
- Department of Medical Neuroscience, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
23
|
Pällmann N, Livgård M, Tesikova M, Zeynep Nenseth H, Akkus E, Sikkeland J, Jin Y, Koc D, Kuzu OF, Pradhan M, Danielsen HE, Kahraman N, Mokhlis HM, Ozpolat B, Banerjee PP, Uren A, Fazli L, Rennie PS, Jin Y, Saatcioglu F. Regulation of the unfolded protein response through ATF4 and FAM129A in prostate cancer. Oncogene 2019; 38:6301-6318. [DOI: 10.1038/s41388-019-0879-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 01/31/2019] [Accepted: 04/10/2019] [Indexed: 12/26/2022]
|
24
|
Zhang DG, Cheng J, Tai ZP, Luo Z. Identification of five genes in endoplasmic reticulum (ER) stress-apoptosis pathways in yellow catfish Pelteobagrus fulvidraco and their transcriptional responses to dietary lipid levels. FISH PHYSIOLOGY AND BIOCHEMISTRY 2019; 45:1117-1127. [PMID: 30847627 DOI: 10.1007/s10695-019-00627-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/26/2019] [Indexed: 06/09/2023]
Abstract
The activating transcription factor 4 (ATF4), DNA damage-inducible transcript 3 (DDIT3), growth arrest, and DNA damage-inducible protein 34 (GADD34), endoplasmic reticulum oxidoreductin 1α (ERO1α), and tumor necrosis factor receptor associated factor 2 (TRAF2) cDNAs were first characterized from yellow catfish Pelteobagrus fulvidraco. Compared to corresponding genes of mammals, all of these proteins shared similar conserved domains. Their mRNAs were widely expressed in various tissues, but at variable levels. Dietary lipid levels did not significantly influence ATF4 mRNA expression. mRNA expression of DDIT3 and GADD34 was highest for fish fed the low-lipid diets and lowest for fish fed middle-lipid diets. The mRNA levels of ERO1α and TRAF2 declined with increasing dietary lipid levels. For the first time, we characterized the full-length cDNA sequences of ATF4, DDIT3, GADD34, ERO1α, and TRAF2 and determined their tissue expression profiles and transcriptional responses to dietary lipid levels, which would contribute to our exploration into their biological functions, and providing new insights on relations between ER stress and lipid metabolism in fish.
Collapse
Affiliation(s)
- Dian-Guang Zhang
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China
| | - Jie Cheng
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China
| | - Zhi-Peng Tai
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China
| | - Zhi Luo
- Fishery College, Huazhong Agricultural University, Wuhan, 43000, China.
| |
Collapse
|
25
|
Abstract
Understanding adipogenesis, the process of adipocyte development, may provide new ways to treat obesity and related metabolic diseases. Adipogenesis is controlled by coordinated actions of lineage-determining transcription factors and epigenomic regulators. Peroxisome proliferator-activated receptor gamma (PPARγ) and C/EBPα are master "adipogenic" transcription factors. In recent years, a growing number of studies have reported the identification of novel transcriptional and epigenomic regulators of adipogenesis. However, many of these novel regulators have not been validated in adipocyte development in vivo and their working mechanisms are often far from clear. In this minireview, we discuss recent advances in transcriptional and epigenomic regulation of adipogenesis, with a focus on factors and mechanisms shared by both white adipogenesis and brown adipogenesis. Studies on the transcriptional regulation of adipogenesis highlight the importance of investigating adipocyte differentiation in vivo rather than drawing conclusions based on knockdown experiments in cell culture. Advances in understanding of epigenomic regulation of adipogenesis have revealed critical roles of histone methylation/demethylation, histone acetylation/deacetylation, chromatin remodeling, DNA methylation, and microRNAs in adipocyte differentiation. We also discuss future research directions that may help identify novel factors and mechanisms regulating adipogenesis.
Collapse
|
26
|
Kuri-Harcuch W, Velez-delValle C, Vazquez-Sandoval A, Hernández-Mosqueira C, Fernandez-Sanchez V. A cellular perspective of adipogenesis transcriptional regulation. J Cell Physiol 2018; 234:1111-1129. [PMID: 30146705 DOI: 10.1002/jcp.27060] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 06/25/2018] [Indexed: 12/19/2022]
Abstract
Adipose cells store lipids in the cytoplasm and signal systemically through secretion of adipokines and other molecules that regulate body energy metabolism. Differentiation of fat cells and its regulation has been the focus of extensive research since the early 1970s. In this review, we had attempted to examine the research bearing on the control of adipose cell differentiation, some of it dating back to the early days when Howard Green and his group described the preadipocyte cell lines 3T3-L1 and 3T3-F442A during 1974-1975. We also concentrated our attention on research published during the last few years, emphasizing data described on transcription factors that regulate adipose differentiation, outside of those that were reported earlier as part of the canonical adipogenic transcriptional cascade, which has been the subject of ample reviews by several groups of researchers. We focused on the studies carried out with the two preadipocyte cell culture models, the 3T3-L1 and 3T3-F442A cells that have provided essential data on adipose biology.
Collapse
Affiliation(s)
- Walid Kuri-Harcuch
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Cristina Velez-delValle
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Alfredo Vazquez-Sandoval
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Claudia Hernández-Mosqueira
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Veronica Fernandez-Sanchez
- Department of Cell Biology, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| |
Collapse
|
27
|
Roumans NJT, Wang P, Vink RG, van Baak MA, Mariman ECM. Combined Analysis of Stress- and ECM-Related Genes in Their Effect on Weight Regain. Obesity (Silver Spring) 2018; 26:492-498. [PMID: 29399976 DOI: 10.1002/oby.22093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 10/14/2017] [Accepted: 10/30/2017] [Indexed: 12/16/2022]
Abstract
OBJECTIVE During weight loss, the volume of adipocytes decreases, leading to stress because of the misfit between the cell contents and the surrounding extracellular matrix (ECM). This stress can be resolved by remodeling the ECM or the restorage of triglycerides within the adipocytes. The objective of this study was to investigate the existence of a connection between stress-related and ECM-related genes that is associated with weight regain. METHODS Thirty-one participants with overweight or obesity followed a 5-week very-low-calorie diet (500 kcal/d) with a subsequent 4-week weight-stable diet (WS), and then an uncontrolled 9-month follow-up. Adipose tissue biopsies were collected for microarray analysis. A correlation and interaction analysis was performed with the weight regain percentage (WR%) ([weight after follow-up - weight after WS] ÷ weight after WS × 100%) by using two gene sets that were previously defined as "stress-related" (n = 107) and "ECM-related" genes (n = 277). RESULTS During WS, a coexpression network of 8 stress-related genes and 15 ECM-related genes correlating with WR% could be constructed, with links to multiple biological processes. Interaction analysis between stress- and ECM-related genes revealed that several gene combinations were highly related to weight regain. CONCLUSIONS Our findings underscore the importance of the connection between stress- and ECM-related genes in the risk for weight regain.
Collapse
Affiliation(s)
- Nadia J T Roumans
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Ping Wang
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
- Department of Clinical Genetics, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Roel G Vink
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Marleen A van Baak
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Edwin C M Mariman
- Department of Human Biology and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
28
|
Yeh KY, Lai CY, Lin CY, Hsu CC, Lo CP, Her GM. ATF4 overexpression induces early onset of hyperlipidaemia and hepatic steatosis and enhances adipogenesis in zebrafish. Sci Rep 2017; 7:16362. [PMID: 29180630 PMCID: PMC5703967 DOI: 10.1038/s41598-017-16587-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 11/13/2017] [Indexed: 02/08/2023] Open
Abstract
Activating transcription factor 4 (ATF4) is constitutively expressed in a variety of tissues, and regulates several pathological features associated with metabolic diseases such as non-alcoholic fatty liver diseases (NAFLD) and obesity. However, the role of ATF4 in animal model systems is poorly understood. To investigate ATF4 functions in zebrafish, we conditionally expressed ATF4 proteins, using a Tet-off transgenic system. We observed early-onset hyperlipidaemia and liver steatosis in ATF4 transgenic zebrafish (ATs) without doxycycline treatment (ATs − Dox). Oil Red O (ORO)-stained signals were predominant in the intravascular blood vessels and liver buds of larval ATs − Dox, indicating that ATF4 functionally promotes lipogenesis. Further, ATF4 overexpression accompanied the stimulation of the unfolded protein response. Therefore, adult ATs − Dox showed increased lipid accumulation, which led, in turn, to liver steatosis. Liver histology and ORO staining of ATs − Dox hepatocytes also indicated oxidative stress and induced NASH-like phenotypes. Moreover, ATF4 overexpression accelerated adipocyte differentiation via CCAAT enhancer binding protein-beta and peroxisome proliferator activated receptor-gamma inducible expression. ATs-Dox zebrafish showed increased weight gain with larger fat pads due to adipocyte hyperplasia. In this study, we report that ATF4 is a potential stimulator of lipid biosynthesis and adipogenesis in zebrafish.
Collapse
Affiliation(s)
- Kun-Yun Yeh
- Division of Hemato-Oncology, Department of Internal Medicine, Chang-Chung Memorial Hospital, 222 Maijin Road, Keelung, 204, Taiwan
| | - Chi-Yu Lai
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, 2, Pei Ning Road, Keelung, 202, Taiwan
| | - Chiu-Ya Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, 2, Pei Ning Road, Keelung, 202, Taiwan
| | - Chia-Chun Hsu
- Department of Radiology, Buddhist Tzu Chi General Hospital, Taichung Branch, No. 66 Fēngxìng Road Section 1, Taichung, 427, Taiwan.,School of Medicine, Tzu Chi University, No.701, Sec. 3, Jhongyang Road, Hualien, 97004, Taiwan
| | - Chung-Ping Lo
- Department of Radiology, Buddhist Tzu Chi General Hospital, Taichung Branch, No. 66 Fēngxìng Road Section 1, Taichung, 427, Taiwan.,School of Medicine, Tzu Chi University, No.701, Sec. 3, Jhongyang Road, Hualien, 97004, Taiwan
| | - Guor Mour Her
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, 2, Pei Ning Road, Keelung, 202, Taiwan.
| |
Collapse
|
29
|
Salvianolic acid B improves glucolipid metabolism by regulating adipogenic transcription factors in mice with diet-induced obesity. JOURNAL OF TRADITIONAL CHINESE MEDICAL SCIENCES 2017. [DOI: 10.1016/j.jtcms.2017.07.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
30
|
Taylor D, Gottlieb RA. Parkin-mediated mitophagy is downregulated in browning of white adipose tissue. Obesity (Silver Spring) 2017; 25:704-712. [PMID: 28240819 PMCID: PMC5373982 DOI: 10.1002/oby.21786] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 01/10/2017] [Accepted: 01/11/2017] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Browning of white adipose tissue (WAT) promotes increased energy expenditure through the action of uncoupling protein 1 (UCP1) and is an attractive target to promote weight loss in obesity. Lowering of mitochondrial membrane potential by UCP1 is uniquely beneficial in this context; in other tissues, reduced membrane potential promotes mitochondrial clearance via mitophagy. It is unknown how parkin-mediated mitophagy is regulated in beige adipocytes. METHODS The relationship between parkin expression and WAT browning was investigated in 3T3-L1 adipocytes and parkin-deficient male C57BL/6 mice in response to pharmacological browning stimuli. RESULTS Rosiglitazone treatment in 3T3-L1 adipocytes promoted mitochondrial biogenesis, UCP1 expression, and mitochondrial uncoupling. Parkin expression was decreased and reduced mitochondrial-associated parkin, and p62 indicated a reduction in mitophagy activity. Parkin overexpression prevented mitochondrial remodeling in response to rosiglitazone. In CL 316,243-treated wild-type mice, decreased parkin expression was observed in subcutaneous inguinal WAT, where UCP1 was strongly induced. CL 316,243 treatment weakly induced UCP1 expression in the gonadal depot, where parkin expression was unchanged. In contrast, parkin-deficient mice exhibited robust UCP1 expression in gonadal WAT following CL 316,243 treatment. CONCLUSIONS WAT browning was associated with a decrease in parkin-mediated mitophagy, and parkin expression antagonized browning of WAT.
Collapse
Affiliation(s)
- David Taylor
- The Cedars-Sinai Heart Institute, Barbra Streisand Women’s Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Roberta A. Gottlieb
- The Cedars-Sinai Heart Institute, Barbra Streisand Women’s Heart Center, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| |
Collapse
|
31
|
Grossman SR, Zhang X, Wang L, Engreitz J, Melnikov A, Rogov P, Tewhey R, Isakova A, Deplancke B, Bernstein BE, Mikkelsen TS, Lander ES. Systematic dissection of genomic features determining transcription factor binding and enhancer function. Proc Natl Acad Sci U S A 2017; 114:E1291-E1300. [PMID: 28137873 PMCID: PMC5321001 DOI: 10.1073/pnas.1621150114] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Enhancers regulate gene expression through the binding of sequence-specific transcription factors (TFs) to cognate motifs. Various features influence TF binding and enhancer function-including the chromatin state of the genomic locus, the affinities of the binding site, the activity of the bound TFs, and interactions among TFs. However, the precise nature and relative contributions of these features remain unclear. Here, we used massively parallel reporter assays (MPRAs) involving 32,115 natural and synthetic enhancers, together with high-throughput in vivo binding assays, to systematically dissect the contribution of each of these features to the binding and activity of genomic regulatory elements that contain motifs for PPARγ, a TF that serves as a key regulator of adipogenesis. We show that distinct sets of features govern PPARγ binding vs. enhancer activity. PPARγ binding is largely governed by the affinity of the specific motif site and higher-order features of the larger genomic locus, such as chromatin accessibility. In contrast, the enhancer activity of PPARγ binding sites depends on varying contributions from dozens of TFs in the immediate vicinity, including interactions between combinations of these TFs. Different pairs of motifs follow different interaction rules, including subadditive, additive, and superadditive interactions among specific classes of TFs, with both spatially constrained and flexible grammars. Our results provide a paradigm for the systematic characterization of the genomic features underlying regulatory elements, applicable to the design of synthetic regulatory elements or the interpretation of human genetic variation.
Collapse
Affiliation(s)
- Sharon R Grossman
- Broad Institute, Cambridge, MA 02142
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Health Sciences and Technology, Harvard Medical School, Boston, MA 02215
| | | | - Li Wang
- Broad Institute, Cambridge, MA 02142
| | - Jesse Engreitz
- Broad Institute, Cambridge, MA 02142
- Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139
| | | | | | - Ryan Tewhey
- Broad Institute, Cambridge, MA 02142
- Faculty of Arts and Sciences Center for Systems Biology, Harvard University, Cambridge, MA 02138
- Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138
| | - Alina Isakova
- Institute of Bioengineering, CH-1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Bart Deplancke
- Institute of Bioengineering, CH-1015 Lausanne, Switzerland
- Swiss Institute of Bioinformatics, CH-1015 Lausanne, Switzerland
| | - Bradley E Bernstein
- Broad Institute, Cambridge, MA 02142
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
- Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114
| | - Tarjei S Mikkelsen
- Broad Institute, Cambridge, MA 02142
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138
| | - Eric S Lander
- Broad Institute, Cambridge, MA 02142;
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139
- Department of Systems Biology, Harvard Medical School, Boston, MA 02215
| |
Collapse
|
32
|
Ayuso M, Fernández A, Núñez Y, Benítez R, Isabel B, Fernández AI, Rey AI, González-Bulnes A, Medrano JF, Cánovas Á, López-Bote CJ, Óvilo C. Developmental Stage, Muscle and Genetic Type Modify Muscle Transcriptome in Pigs: Effects on Gene Expression and Regulatory Factors Involved in Growth and Metabolism. PLoS One 2016; 11:e0167858. [PMID: 27936208 PMCID: PMC5148031 DOI: 10.1371/journal.pone.0167858] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 11/21/2016] [Indexed: 01/08/2023] Open
Abstract
Iberian pig production includes purebred (IB) and Duroc-crossbred (IBxDU) pigs, which show important differences in growth, fattening and tissue composition. This experiment was conducted to investigate the effects of genetic type and muscle (Longissimus dorsi (LD) vs Biceps femoris (BF)) on gene expression and transcriptional regulation at two developmental stages. Nine IB and 10 IBxDU piglets were slaughtered at birth, and seven IB and 10 IBxDU at four months of age (growing period). Carcass traits and LD intramuscular fat (IMF) content were measured. Muscle transcriptome was analyzed on LD samples with RNA-Seq technology. Carcasses were smaller in IB than in IBxDU neonates (p < 0.001), while growing IB pigs showed greater IMF content (p < 0.05). Gene expression was affected (p < 0.01 and Fold change > 1.5) by the developmental stage (5,812 genes), muscle type (135 genes), and genetic type (261 genes at birth and 113 at growth). Newborns transcriptome reflected a highly proliferative developmental stage, while older pigs showed upregulation of catabolic and muscle functioning processes. Regarding the genetic type effect, IBxDU newborns showed enrichment of gene pathways involved in muscle growth, in agreement with the higher prenatal growth observed in these pigs. However, IB growing pigs showed enrichment of pathways involved in protein deposition and cellular growth, supporting the compensatory gain experienced by IB pigs during this period. Moreover, newborn and growing IB pigs showed more active glucose and lipid metabolism than IBxDU pigs. Moreover, LD muscle seems to have more active muscular and cell growth, while BF points towards lipid metabolism and fat deposition. Several regulators controlling transcriptome changes in both genotypes were identified across muscles and ages (SIM1, PVALB, MEFs, TCF7L2 or FOXO1), being strong candidate genes to drive expression and thus, phenotypic differences between IB and IBxDU pigs. Many of the identified regulators were known to be involved in muscle and adipose tissues development, but others not previously associated with pig muscle growth were also identified, as PVALB, KLF1 or IRF2. The present study discloses potential molecular mechanisms underlying phenotypic differences observed between IB and IBxDU pigs and highlights candidate genes implicated in these molecular mechanisms.
Collapse
Affiliation(s)
- Miriam Ayuso
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Yolanda Núñez
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| | - Rita Benítez
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| | - Beatriz Isabel
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Ana I. Rey
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | | | - Juan F. Medrano
- Department of Animal Science, University of California Davis, Davis, California, United States of America
| | - Ángela Cánovas
- Department of Animal Science, University of California Davis, Davis, California, United States of America
| | - Clemente J. López-Bote
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Cristina Óvilo
- Departamento de Mejora Genética Animal, INIA, Madrid, Spain
| |
Collapse
|
33
|
Dusseault J, Li B, Haider N, Goyette MA, Côté JF, Larose L. Nck2 Deficiency in Mice Results in Increased Adiposity Associated With Adipocyte Hypertrophy and Enhanced Adipogenesis. Diabetes 2016; 65:2652-66. [PMID: 27325288 DOI: 10.2337/db15-1559] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 06/09/2016] [Indexed: 11/13/2022]
Abstract
Obesity results from an excessive expansion of white adipose tissue (WAT) from hypertrophy of preexisting adipocytes and enhancement of precursor differentiation into mature adipocytes. We report that Nck2-deficient mice display progressive increased adiposity associated with adipocyte hypertrophy. A negative relationship between the expression of Nck2 and WAT expansion was recapitulated in humans such that reduced Nck2 protein and mRNA levels in human visceral WAT significantly correlate with the degree of obesity. Accordingly, Nck2 deficiency promotes an adipogenic program that not only enhances adipocyte differentiation and lipid droplet formation but also results in dysfunctional elevated lipogenesis and lipolysis activities in mouse WAT as well as in stromal vascular fraction and 3T3-L1 preadipocytes. We provide strong evidence to support that through a mechanism involving primed PERK activation and signaling, Nck2 deficiency in adipocyte precursors is associated with enhanced adipogenesis in vitro and adiposity in vivo. Finally, in agreement with elevated circulating lipids, Nck2-deficient mice develop glucose intolerance, insulin resistance, and hepatic steatosis. Taken together, these findings reveal that Nck2 is a novel regulator of adiposity and suggest that Nck2 is important in limiting WAT expansion and dysfunction in mice and humans.
Collapse
Affiliation(s)
- Julie Dusseault
- Department of Medicine, McGill University, and McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | - Bing Li
- Department of Medicine, McGill University, and McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | - Nida Haider
- Department of Medicine, McGill University, and McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | - Marie-Anne Goyette
- Institut de Recherches Cliniques de Montréal (Université de Montréal), Montreal, Quebec, Canada
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal (Université de Montréal), Montreal, Quebec, Canada
| | - Louise Larose
- Department of Medicine, McGill University, and McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| |
Collapse
|
34
|
Chen H, Yuan R, Zhang Y, Zhang X, Chen L, Zhou X, Yuan Z, Nie Y, Li M, Mo D, Chen Y. ATF4 regulates SREBP1c expression to control fatty acids synthesis in 3T3-L1 adipocytes differentiation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2016; 1859:1459-1469. [PMID: 27452504 DOI: 10.1016/j.bbagrm.2016.07.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 07/07/2016] [Accepted: 07/20/2016] [Indexed: 11/26/2022]
Abstract
Activating transcription factor 4 (ATF4), which is highly expressed in 3T3-L1 adipocytes after adipogenic induction, is essential for adipocytes differentiation. ATF4 also plays a vital role in regulating fatty acids biosynthesis, whereas the detailed mechanism of this process is still unclear. Here we demonstrated that siRNA-based ATF4 depletion in 3T3-L1 adipocytes significantly reduced the accumulation of fatty acids and triglycerides. Moreover, SREBP1c protein, which is an important transcription factor of lipogenesis, appreciably decreased while Srebp1c mRNA increased. Then we identified that ATF4 could maintain SREBP1c protein stability by directly activating the expression of USP7 which deubiquitinates SREBP1c and increases its protein content in cell. Besides, USP7 could restore the synthesis of fatty acids and triglycerides in the absence of ATF4. On the other hand, we found that ATF4 might inhibit the transcription of Srebp1c through TRB3, which is repressed by IBMX and DEX during early adipogenesis. Thus, our data indicate that ATF4 regulates SREBP1c expression to control fatty acids synthesis.
Collapse
Affiliation(s)
- Hu Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Renqiang Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ying Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xumeng Zhang
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Luxi Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xingyu Zhou
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Zhuning Yuan
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yaping Nie
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ming Li
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Delin Mo
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| | - Yaosheng Chen
- State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
35
|
Wang M, Kaufman RJ. Protein misfolding in the endoplasmic reticulum as a conduit to human disease. Nature 2016; 529:326-35. [PMID: 26791723 DOI: 10.1038/nature17041] [Citation(s) in RCA: 1075] [Impact Index Per Article: 134.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 11/11/2015] [Indexed: 12/18/2022]
Abstract
In eukaryotic cells, the endoplasmic reticulum is essential for the folding and trafficking of proteins that enter the secretory pathway. Environmental insults or increased protein synthesis often lead to protein misfolding in the organelle, the accumulation of misfolded or unfolded proteins - known as endoplasmic reticulum stress - and the activation of the adaptive unfolded protein response to restore homeostasis. If protein misfolding is not resolved, cells die. Endoplasmic reticulum stress and activation of the unfolded protein response help to determine cell fate and function. Furthermore, endoplasmic reticulum stress contributes to the aetiology of many human diseases.
Collapse
Affiliation(s)
- Miao Wang
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, California 92037, USA
| |
Collapse
|
36
|
Cohen DM, Won KJ, Nguyen N, Lazar MA, Chen CS, Steger DJ. ATF4 licenses C/EBPβ activity in human mesenchymal stem cells primed for adipogenesis. eLife 2015; 4:e06821. [PMID: 26111340 PMCID: PMC4501333 DOI: 10.7554/elife.06821] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 06/25/2015] [Indexed: 12/24/2022] Open
Abstract
A well-established cascade of transcription factor (TF) activity orchestrates adipogenesis in response to chemical cues, yet how cell-intrinsic determinants of differentiation such as cell shape and/or seeding density inform this transcriptional program remain enigmatic. Here, we uncover a novel mechanism licensing transcription in human mesenchymal stem cells (hMSCs) adipogenically primed by confluence. Prior to adipogenesis, confluency promotes heterodimer recruitment of the bZip TFs C/EBPβ and ATF4 to a non-canonical C/EBP DNA sequence. ATF4 depletion decreases both cell-density-dependent transcription and adipocyte differentiation. Global profiling in hMSCs and a novel cell-free assay reveals that ATF4 requires C/EBPβ for genomic binding at a motif distinct from that bound by the C/EBPβ homodimer. Our observations demonstrate that C/EBPβ bridges the transcriptional programs in naïve, confluent cells and early differentiating pre-adipocytes. Moreover, they suggest that homo- and heterodimer formation poise C/EBPβ to execute diverse and stage-specific transcriptional programs by exploiting an expanded motif repertoire.
Collapse
Affiliation(s)
- Daniel M Cohen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Kyoung-Jae Won
- The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Nha Nguyen
- The Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Mitchell A Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Boston University, Boston, United States
| | - David J Steger
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States
| |
Collapse
|
37
|
Cho YM, Kwak SN, Joo NS, Kim DH, Lee AH, Kim KS, Seo JB, Jeong SW, Kwon OJ. X-box binding protein 1 is a novel key regulator of peroxisome proliferator-activated receptor γ2. FEBS J 2014; 281:5132-46. [PMID: 25223794 DOI: 10.1111/febs.13052] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 08/10/2014] [Accepted: 09/12/2014] [Indexed: 01/08/2023]
Abstract
X-box binding protein 1 (XBP1), a transcription factor of the unfolded protein response, plays various roles in many biological processes. We examined its pro-adipogenic activity and target genes during adipogenic differentiation in wild-type and genetically modified 3T3-L1 cells. Signalling pathways that contribute to Xbp1 mRNA splicing, and the correlation of the transcriptionally active XBP1 isoform (XBP1s) level with body mass index and the level of peroxisome proliferator-activated receptor γ2 (PPARγ2) in human adipose tissues were also examined. The mRNA and nuclear protein expression levels of XBP1s increased immediately following hormonal induction of adipogenesis, reaching a peak at 6 h. Results from cDNA microarray and gene expression analyses using genetically modified cells indicated that PPARγ2 was a principal target of XBP1s. The XBP1s-specific binding motif, which is distinct from the CCAAT/enhancer-binding protein α binding site, was identified in the PPARγ2 promoter by site-directed mutagenesis. Fetal bovine serum, insulin, 3-isobutyl-1-methylxanthine and dexamethasone contributed independently to Xbp1 mRNA splicing. In human subcutaneous adipose tissues, the levels of both Xbp1s and Pparγ2 mRNA increased proportionally with body mass index, and there was a significant positive correlation between the two genes. These data suggest for the first time that positive regulation of PPARγ2 is a principal mechanism of XBP1s-mediated adipogenesis in 3T3-L1 cells.
Collapse
Affiliation(s)
- Yoon Mi Cho
- Department of Biochemistry, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|