1
|
Reynolds AZ, Niedbalski SD. Sex-biased gene regulation varies across human populations as a result of adaptive evolution. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2024; 183:e24888. [PMID: 38100225 PMCID: PMC11279473 DOI: 10.1002/ajpa.24888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 11/14/2023] [Accepted: 11/28/2023] [Indexed: 03/03/2024]
Abstract
OBJECTIVES Studies of human sexual dimorphism and gender disparities in health focus on ostensibly universal molecular sex differences, such as sex chromosomes and circulating hormone levels, while ignoring the extraordinary diversity in biology, behavior, and culture acquired by different human populations over their unique evolutionary histories. MATERIALS AND METHODS Using RNA-Seq data and whole genome sequences from 1000G and HGDP, we investigate variation in sex-biased gene expression across 11 human populations and test whether population-level variation in sex-biased expression may have resulted from adaptive evolution in regions containing sex-specific regulatory variants. RESULTS We find that sex-biased gene expression in humans is highly variable, mostly population-specific, and demonstrates between population reversals. Expression quantitative trait locus mapping reveals sex-specific regulatory regions with evidence of recent positive natural selection, suggesting that variation in sex-biased expression may have evolved as an adaptive response to ancestral environments experienced by human populations. DISCUSSION These results indicate that sex-biased gene expression is more flexible than previously thought and is not generally shared among human populations. Instead, molecular phenotypes associated with sex depend on complex interactions between population-specific molecular evolution and physiological responses to contemporary socioecologies.
Collapse
Affiliation(s)
- Adam Z. Reynolds
- Department of Anthropology, University of New Mexico, Albuquerque, NM
| | | |
Collapse
|
2
|
Qiu S, Sun Y, Guo J, Zhang Y, Hu Y. Genome-wide analysis reveals extensive genetic overlap between childhood phenotypes and later-life type 2 diabetes. Comput Biol Med 2024; 171:108065. [PMID: 38387379 DOI: 10.1016/j.compbiomed.2024.108065] [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: 11/12/2023] [Revised: 12/26/2023] [Accepted: 01/27/2024] [Indexed: 02/24/2024]
Abstract
Observational studies have indicated a potential influence of childhood phenotypes on the later development of type 2 diabetes (T2D). However, the underlying biological mechanisms remain unclear. In this study, we conducted a comprehensive genome-wide analysis to investigate the shared genetic architecture and genetic loci between nine childhood phenotypes (N = 4202-620,26) and later-life T2D (N = 80,154) using genetic correlation, mendelian randomization (MR), and conjunctional false discovery rate (conjFDR) statistical frameworks. Our findings demonstrated substantial genetic correlations and pleiotropic enrichment between childhood obesity, body mass index (BMI), systolic blood pressure (SBP), diastolic blood pressure (DBP), and later-life T2D. Childhood obesity exhibited a significant association with increased later-life T2D risk through 10 mediators, 6 of which were adulthood obesity-related phenotypes. Additionally, we identified 69, 83, 3, 5, 10, 5, 3, and 7 loci shared between childhood obesity, BMI, SBP, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), triglycerides (TG), apolipoprotein A-I (ApoA-I), apolipoprotein B (ApoB), and T2D at conjFDR <0.05, with the majority of these loci being novel discoveries. Overall, our study reveals extensive genetic overlap between childhood obesity-related phenotypes and T2D with concordant effect directions, shedding new light on variants and phenotypes with lifelong effects.
Collapse
Affiliation(s)
- Shizheng Qiu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China
| | - Yige Sun
- Department of Radiology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jiahe Guo
- School of Future Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yu Zhang
- Beidahuang Industry Group General Hospital, Harbin, 150088, China.
| | - Yang Hu
- School of Computer Science and Technology, Harbin Institute of Technology, Harbin, Heilongjiang, 150001, China.
| |
Collapse
|
3
|
Hwang MS, Park J, Ham Y, Lee IH, Chun KH. Roles of Protein Post-Translational Modifications During Adipocyte Senescence. Int J Biol Sci 2023; 19:5245-5256. [PMID: 37928271 PMCID: PMC10620833 DOI: 10.7150/ijbs.86404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 09/27/2023] [Indexed: 11/07/2023] Open
Abstract
Adipocytes are adipose tissues that supply energy to the body through lipids. The two main types of adipocytes comprise white adipocytes (WAT) that store energy, and brown adipocytes (BAT), which generate heat by burning stored fat (thermogenesis). Emerging evidence indicates that dysregulated adipocyte senescence may disrupt metabolic homeostasis, leading to various diseases and aging. Adipocytes undergo senescence via irreversible cell-cycle arrest in response to DNA damage, oxidative stress, telomere dysfunction, or adipocyte over-expansion upon chronic lipid accumulation. The amount of detectable BAT decreases with age. Activation of cell cycle regulators and dysregulation of adipogenesis-regulating factors may constitute a molecular mechanism that accelerates adipocyte senescence. To better understand the regulation of adipocyte senescence, the effects of post-translational modifications (PTMs), is essential for clarifying the activity and stability of these proteins. PTMs are covalent enzymatic protein modifications introduced following protein biosynthesis, such as phosphorylation, acetylation, ubiquitination, or glycosylation. Determining the contribution of PTMs to adipocyte senescence may identify new therapeutic targets for the regulation of adipocyte senescence. In this review, we discuss a conceptual case in which PTMs regulate adipocyte senescence and explain the mechanisms underlying protein regulation, which may lead to the development of effective strategies to combat metabolic diseases.
Collapse
Affiliation(s)
- Min-Seon Hwang
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jingyeong Park
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Yunha Ham
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - In Hye Lee
- Department of Life Science, College of Natural Science, Ewha Womans University, 52 Ewhayeodae-Gil, Seodaemun-gu, Seoul, 03760, Republic of Korea
| | - Kyung-Hee Chun
- Department of Biochemistry & Molecular Biology, Graduate School of Medical Science, Brain Korea 21 Project, Institute of Genetic Science, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| |
Collapse
|
4
|
Sun J, Jin X, Zhang X, Zhang B. HMGA2 knockdown alleviates the progression of nonalcoholic fatty liver disease (NAFLD) by downregulating SNAI2 expression. Cell Signal 2023:110741. [PMID: 37268162 DOI: 10.1016/j.cellsig.2023.110741] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/23/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a complex disease that is considered as the next major health epidemic with alarmingly increasing global prevalence. To explore the pathogenesis of NAFLD, data from GSE118892 were analyzed. High mobility group AT-hook 2 (HMGA2), a member of the high mobility group family, is declined in liver tissues of NAFLD rats. However, its role in NAFLD remains unknown. This study attempted to identify the multiple roles of HMGA2 in NAFLD process. NAFLD was induced in rats using a high-fat diet (HFD). In vivo, HMGA2 knockdown using adenovirus system attenuated liver injury and liver lipid deposition, accompanied by decreased NAFLD score, increased liver function, and decreased CD36 and FAS, indicating the deceleration of NAFLD progression. Moreover, HMGA2 knockdown restrained liver inflammation by decreasing the expression of related inflammatory factors. Importantly, HMGA2 knockdown attenuated liver fibrosis via downregulating the expression of fibrous proteins, and inhibiting the activation of TGF-β1/SMAD signaling pathway. In vitro, HMGA2 knockdown relieved palmitic acid (PA)-induced hepatocyte injury and attenuated TGF-β1-induced liver fibrosis, consistent with in vivo findings. Strikingly, HMGA2 activated the transcription of SNAI2, which was evidenced by the dual luciferase assays. Moreover, HMGA2 knockdown largely downregulated SNAI2 levels. Indeed, SNAI2 overexpression effectively blocked the inhibitory effect of HMGA2 knockdown on NAFLD. Totally, our findings reveal that HMGA2 knockdown alleviates the progression of NAFLD by directly regulating the transcription of SNAI2. HMGA2 inhibition may emerge as a potential therapeutic target for NAFLD.
Collapse
Affiliation(s)
- Jing Sun
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China.
| | - Xiuli Jin
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Xinhe Zhang
- Department of Gastroenterology, the First Hospital of China Medical University, Shenyang, Liaoning Province, People's Republic of China
| | - Birong Zhang
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff University, Cardiff, UK
| |
Collapse
|
5
|
Roy D, Purohit P, Khokhar M, Modi A, Shukla RKG, Chaudhary R, Sankanagoudar S, Sharma P. Analyzing the Association of Visceral Adipose Tissue Growth Differentiation Factor-15 and MicroRNA in Type 2 Diabetes Mellitus. J Obes Metab Syndr 2023; 32:64-76. [PMID: 36918405 PMCID: PMC10088550 DOI: 10.7570/jomes22010] [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] [Received: 02/12/2022] [Revised: 04/07/2022] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Background Growth differentiation factor-15 (GDF-15) is involved in insulin resistance and diabetes. In this study, we determine the associations of GDF-15 with miR-181b-5p, miR-330-3p, mothers against decapentaplegic homolog 7 (SMAD7), and insulin resistance in visceral adipose tissue (VAT) and peripheral blood mononuclear cells (PBMCs) in type 2 diabetes mellitus (T2DM) patients. Methods Sixty patients, equally divided into those with T2DM and non-diabetic controls, were recruited for gene expression analysis. Protein-protein interaction (STRING), target prediction (miRNet), and functional enrichment were conducted accordingly. Results Our study showed that VAT and PBMCs had similar expression profiles, where GDF-15 and miR-181b-5p were upregulated, whereas SMAD7 and miR-330-3p were downregulated. Serum GDF-15 could differentiate between T2DM and non-diabetic patients (P<0.001). Target prediction revealed a microRNA (miRNA)-messenger RNA regulatory network, transcription factors, and functional enrichment for the miRNA that suggested involvement in T2DM pathogenesis. Conclusion VAT GDF-15 is associated with insulin resistance and is possibly regulated by miR-181b-5p, miR-330-3p, and SMAD7 in T2DM.
Collapse
Affiliation(s)
- Dipayan Roy
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
- Indian Institute of Technology (ITT)-Madras, Chennai, India
- School of Humanities, Indira Gandhi National Open University (IGNOU), New Delhi, India
| | - Purvi Purohit
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Manoj Khokhar
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | - Anupama Modi
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | | | - Ramkaran Chaudhary
- Department of General Surgery, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| | | | - Praveen Sharma
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), Jodhpur, India
| |
Collapse
|
6
|
Dapas M, Dunaif A. Deconstructing a Syndrome: Genomic Insights Into PCOS Causal Mechanisms and Classification. Endocr Rev 2022; 43:927-965. [PMID: 35026001 PMCID: PMC9695127 DOI: 10.1210/endrev/bnac001] [Citation(s) in RCA: 87] [Impact Index Per Article: 43.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Indexed: 01/16/2023]
Abstract
Polycystic ovary syndrome (PCOS) is among the most common disorders in women of reproductive age, affecting up to 15% worldwide, depending on the diagnostic criteria. PCOS is characterized by a constellation of interrelated reproductive abnormalities, including disordered gonadotropin secretion, increased androgen production, chronic anovulation, and polycystic ovarian morphology. It is frequently associated with insulin resistance and obesity. These reproductive and metabolic derangements cause major morbidities across the lifespan, including anovulatory infertility and type 2 diabetes (T2D). Despite decades of investigative effort, the etiology of PCOS remains unknown. Familial clustering of PCOS cases has indicated a genetic contribution to PCOS. There are rare Mendelian forms of PCOS associated with extreme phenotypes, but PCOS typically follows a non-Mendelian pattern of inheritance consistent with a complex genetic architecture, analogous to T2D and obesity, that reflects the interaction of susceptibility genes and environmental factors. Genomic studies of PCOS have provided important insights into disease pathways and have indicated that current diagnostic criteria do not capture underlying differences in biology associated with different forms of PCOS. We provide a state-of-the-science review of genetic analyses of PCOS, including an overview of genomic methodologies aimed at a general audience of non-geneticists and clinicians. Applications in PCOS will be discussed, including strengths and limitations of each study. The contributions of environmental factors, including developmental origins, will be reviewed. Insights into the pathogenesis and genetic architecture of PCOS will be summarized. Future directions for PCOS genetic studies will be outlined.
Collapse
Affiliation(s)
- Matthew Dapas
- Department of Human Genetics, University of Chicago, Chicago, IL, USA
| | - Andrea Dunaif
- Division of Endocrinology, Diabetes and Bone Disease, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
7
|
Concistrè A, Petramala L, Circosta F, Romagnoli P, Soldini M, Bucci M, De Cesare D, Cavallaro G, De Toma G, Cipollone F, Letizia C. Analysis of the miRNA expression from the adipose tissue surrounding the adrenal neoplasia. Front Cardiovasc Med 2022; 9:930959. [PMID: 35966515 PMCID: PMC9366211 DOI: 10.3389/fcvm.2022.930959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/01/2022] [Indexed: 12/04/2022] Open
Abstract
Background Primary aldosteronism (PA) is characterized by several metabolic changes such as insulin resistance, metabolic syndrome, and adipose tissue (AT) inflammation. Mi(cro)RNAs (miRNAs) are a class of non-coding small RNA molecules known to be critical regulators in several cellular processes associated with AT dysfunction. The aim of this study was to evaluate the expression of some miRNAs in visceral and subcutaneous AT in patients undergoing adrenalectomy for aldosterone-secreting adrenal adenoma (APA) compared to the samples of AT obtained in patients undergoing adrenalectomy for non-functioning adrenal mass (NFA). Methods The quantitative expression of selected miRNA using real-time PCR was analyzed in surrounding adrenal neoplasia, peri-renal, and subcutaneous AT samples of 16 patients with adrenalectomy (11 patients with APA and 5 patients with NFA). Results Real-time PCR cycles for miRNA-132, miRNA-143, and miRNA-221 in fat surrounding adrenal neoplasia and in peri-adrenal AT were significantly higher in APA than in patients with NFA. Unlike patients with NFA, miRNA-132, miRNA-143, miRNA-221, and miRNA-26b were less expressed in surrounding adrenal neoplasia AT compared to subcutaneous AT in patients with APA. Conclusion This study, conducted on tissue expression of miRNAs, highlights the possible pathophysiological role of some miRNAs in determining the metabolic alterations in patients with PA.
Collapse
Affiliation(s)
- Antonio Concistrè
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Luigi Petramala
- Department of Translational and Precision Medicine, “Sapienza” University of Rome, Rome, Italy
| | - Francesco Circosta
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Priscilla Romagnoli
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Maurizio Soldini
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, “Sapienza” University of Rome, Rome, Italy
| | - Marco Bucci
- Department of Medicine and Aging Sciences, University “Gabriele d'Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Domenico De Cesare
- Department of Medicine and Aging Sciences, University “Gabriele d'Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Giuseppe Cavallaro
- Department of Surgery “Pietro Valdoni, ” “Sapienza” University of Rome, Rome, Italy
| | - Giorgio De Toma
- Department of Surgery “Pietro Valdoni, ” “Sapienza” University of Rome, Rome, Italy
| | - Francesco Cipollone
- Department of Medicine and Aging Sciences, University “Gabriele d'Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Claudio Letizia
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, “Sapienza” University of Rome, Rome, Italy
- *Correspondence: Claudio Letizia
| |
Collapse
|
8
|
Chen D, Wu H, Wang X, Huang T, Jia J. Shared Genetic Basis and Causal Relationship Between Television Watching, Breakfast Skipping and Type 2 Diabetes: Evidence From a Comprehensive Genetic Analysis. Front Endocrinol (Lausanne) 2022; 13:836023. [PMID: 35399945 PMCID: PMC8988136 DOI: 10.3389/fendo.2022.836023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/01/2022] [Indexed: 11/13/2022] Open
Abstract
Background Epidemiological investigations have established unhealthy lifestyles, such as excessive leisurely sedentary behavior (especially TV/television watching) and breakfast skipping, increase the risk of type 2 diabetes (T2D), but the causal relationship is unclear. We aimed to understand how single nucleotide variants contribute to the co-occurrence of unhealthy lifestyles and T2D, thereby providing meaningful insights into disease mechanisms. Methods Combining summary statistics from genome-wide association studies (GWAS) on TV watching (N = 422218), breakfast skipping (N = 193860) and T2D (N = 159208) in European pedigrees, we conducted comprehensive pairwise genetic analysis, including high-definition likelihood (HDL-method), cross-phenotype association studies (CPASSOC), GWAS-eQTL colocalization analysis and transcriptome-wide association studies (TWAS), to understand the genetic overlap between them. We also performed bidirectional two-sample Mendelian randomization (MR) analysis for causal inference using genetic instrumental variables, and two-step MR mediation analysis was used to assess any effects explained by body mass index, lipid traits and glycemic traits. Results HDL-method showed that T2D shared a strong genetic correlation with TV watching (rg = 0.26; P = 1.63×10-29) and skipping breakfast (rg = 0.15; P =2.02×10-6). CPASSOC identifies eight independent SNPs shared between T2D and TV watching, including one novel shared locus. TWAS and CPASSOC showed that shared genes were enriched in lung, esophageal, adipose, and thyroid tissues and highlighted potential shared regulatory pathways for lipoprotein metabolism, pancreatic β-cell function, cellular senescence and multi-mediator factors. MR showed TV watching had a causal effect on T2D (βIVW = 0.629, PIVW = 1.80×10-10), but no significant results were observed between breakfast skipping and T2D. Mediation analysis provided evidence that body mass index, fasting glucose, hemoglobin A1c and high-density lipoprotein are potential factors that mediate the causal relationship between TV and T2D. Conclusions Our findings provide strong evidence of shared genetics and causation between TV watching and T2D and facilitate our identification of common genetic architectures shared between them.
Collapse
Affiliation(s)
- Dongze Chen
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Hanyu Wu
- Department of Bioinformatics, School of Life Science, Peking University, Beijing, China
| | - Xinpei Wang
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Tao Huang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University, Beijing, China
- Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Ministry of Education, Beijing, China
| | - Jinzhu Jia
- Department of Biostatistics, School of Public Health, Peking University, Beijing, China
- Center for Statistical Science, Peking University, Beijing, China
| |
Collapse
|
9
|
Zhao F, Wu L, Wang Q, Zhao X, Chen T, Yin C, Yan L, Yang X. Insulin-like growth factor 2 mRNA-binding protein 2-regulated alternative splicing of nuclear factor 1 C-type causes excessive granulosa cell proliferation in polycystic ovary syndrome. Cell Prolif 2022; 55:e13216. [PMID: 35293050 PMCID: PMC9055906 DOI: 10.1111/cpr.13216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 02/12/2022] [Indexed: 12/02/2022] Open
Abstract
Objectives Polycystic ovary syndrome (PCOS) is a common reproductive endocrine disorder. Insulin‐like growth factor 2 mRNA‐binding protein 2 (IGF2BP2) serves as an HMGA2 target gene to promote the proliferation of granulosa cells (GCs). However, it is still unclear whether IGF2BP2 participates in the pathogenesis of PCOS as RNA binding protein (RBP). In this study, we aimed to elucidate IGF2BP2‐interacting transcripts, global transcriptome together with alternative splicing in GCs to eventually uncover potential mechanisms of PCOS pathogenesis. Materials and Methods The expression of IGF2BP2 in GCs from PCOS patients was detected using quantitative reverse transcription PCR (RT‐qPCR) and western blot. We captured IGF2BP2‐interacting transcripts, global transcriptome together with alternative splicing by RNA immunoprecipitation sequencing (RIP‐seq) and RNA sequencing (RNA‐seq). KGN cells transfected with IGF2BP2 overexpressing plasmids and nuclear factor 1 C‐type (NFIC) siRNAs, were applied to CCK‐8, EdU and TUNEL assays. Results IGF2BP2 was highly expressed in GCs from PCOS patients. As an RBP, it preferentially bound to the 3′and 5′UTRs of mRNAs with GGAC motif and a newly found GAAG motif. The overexpression of IGF2BP2 changed the transcriptome profile of KGN cells. IGF2BP2 functioned to regulate alternative splicing events and promote cell proliferation through inhibiting exon skipping events of NFIC. Conclusion In conclusion, we demonstrated that IGF2BP2 promotes GC proliferation via regulating alternative splicing of NFIC in PCOS. The findings help to better understand the roles of IGF2BP2 in the pathogenesis of PCOS.
Collapse
Affiliation(s)
- Feiyan Zhao
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China.,Department of Human Reproductive Medicine, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Liang Wu
- Reproductive Medical Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China.,Henan Key Laboratory of Reproduction and Genetics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, People's Republic of China
| | - Qin Wang
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China.,Department of Human Reproductive Medicine, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
| | - Xuehan Zhao
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China.,Department of Human Reproductive Medicine, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China.,State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China
| | - Tong Chen
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China.,Department of Human Reproductive Medicine, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
| | - Chenghong Yin
- Department of Human Reproductive Medicine, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China.,Department of Internal Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Long Yan
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, People's Republic of China.,University of Chinese Academy of Sciences, Beijing, People's Republic of China.,Department for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, People's Republic of China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, People's Republic of China
| | - Xiaokui Yang
- Department of Human Reproductive Medicine, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, People's Republic of China.,Department of Human Reproductive Medicine, Beijing Maternal and Child Health Care Hospital, Beijing, People's Republic of China
| |
Collapse
|
10
|
Pan X, Zhang M, Tian A, Chen L, Sun Z, Wang L, Chen P. Exploring the genetic correlation between obesity-related traits and regional brain volumes: Evidence from UK Biobank cohort. Neuroimage Clin 2022; 33:102870. [PMID: 34872017 PMCID: PMC8648807 DOI: 10.1016/j.nicl.2021.102870] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 10/27/2021] [Accepted: 10/28/2021] [Indexed: 01/21/2023]
Abstract
OBJECTIVE To determine whether there is a correlation between obesity-related variants and regional brain volumes. METHODS Based on a mixed linear model (MLM), we analyzed the association between 1,498 obesity-related SNPs in the GWAS Catalog and 164 regional brain volumes from 29,420 participants (discovery cohort N = 19,997, validation cohort N = 9,423) in UK Biobank. The statistically significant brain regions in association analysis were classified into 6 major neural networks (dopamine (DA) motive system, central autonomic network (CAN), cognitive emotion regulation, visual object recognition network, auditory object recognition network, and sensorimotor system). We summarized the association between obesity-related variants (metabolically healthy obesity variants, metabolically unhealthy obesity variants, and unclassified obesity-related variants) and neural networks. RESULTS From association analysis, we determined that 17 obesity-related SNPs were associated with 51 regional brain volumes. Several single SNPs (e.g., rs13107325-T (SLC39A8), rs1876829-C (CRHR1), and rs1538170-T (CENPW)) were associated with multiple regional brain volumes. In addition, several single brain regions (e.g., the white matter, the grey matter in the putamen, subcallosal cortex, and insular cortex) were associated with multiple obesity-related variants. The metabolically healthy obesity variants were mainly associated with the regional brain volumes in the DA motive system, sensorimotor system and cognitive emotion regulation neural networks, while metabolically unhealthy obesity variants were mainly associated with regional brain volumes in the CAN and total tissue volumes. In addition, unclassified obesity-related variants were mainly associated with auditory object recognition network and total tissue volumes. The results of MeSH (medical subject headings) enrichment analysis showed that obesity genes associated with brain structure pointed to the functional relatedness with 5-Hydroxytryptamine receptor 4 (5-HT4), growth differentiation factor 5 (GDF5), and high mobility group protein AT-hook 2 (HMGA2 protein). CONCLUSION In summary, we found that obesity-related variants were associated with different brain volume measures. On the basis of the multiple SNPs, we found that metabolically healthy and unhealthy obesity-related SNPs were associated with different brain neural networks. Based on our enrichment analysis, modifications of the 5-HT4 pathway might be a promising therapeutic strategy for obesity.
Collapse
Affiliation(s)
- Xingchen Pan
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China; Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Miaoran Zhang
- Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Aowen Tian
- Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Lanlan Chen
- School of Clinical Medicine, Jilin University, Changchun, 130000, China
| | - Zewen Sun
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China
| | - Liying Wang
- Department of Molecular Biology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| | - Peng Chen
- Department of Genetics, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China; Department of Pathology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
11
|
Tasin FR, Ahmed A, Halder D, Mandal C. On-going consequences of in utero exposure of Pb: An epigenetic perspective. J Appl Toxicol 2022; 42:1553-1569. [PMID: 35023172 DOI: 10.1002/jat.4287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 12/23/2021] [Accepted: 01/01/2022] [Indexed: 11/08/2022]
Abstract
Epigenetic modifications by toxic heavy metals are one of the intensively investigated fields of modern genomic research. Among a diverse group of heavy metals, lead (Pb) is an extensively distributed toxicant causing an immense number of abnormalities in the developing fetus via a wide variety of epigenetic changes. As a divalent cation, Pb can readily cross the placental membrane and the fetal blood brain barrier leading to far-reaching alterations in DNA methylation patterns, histone protein modifications and micro-RNA expression. Over recent years, several human cohorts and animal model studies have documented hyper- and hypo-methylation of developmental genes along with altered DNA methyl-transferase expression by in utero Pb exposure in a dose-, duration- and sex-dependent manner. Modifications in the expression of specific histone acetyltransferase enzymes along with histone acetylation and methylation levels have been reported in rodent and murine models. Apart from these, down-regulation and up-regulation of certain microRNAs crucial for fetal development have been shown to be associated with in utero Pb exposure in human placenta samples. All these modifications in the developing fetus during the prenatal and perinatal stages reportedly caused severe abnormalities in early or adult age, such as - impaired growth, obesity, autism, diabetes, cardiovascular diseases, risks of cancer development and Alzheimer's disease. In this review, currently available information on Pb-mediated alterations in the fetal epigenome is summarized. Further research on Pb-induced epigenome modification will help to understand the mechanisms in detail and will enable us to formulate safety guidelines for pregnant women and developing children.
Collapse
Affiliation(s)
- Fahim Rejanur Tasin
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Asif Ahmed
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Debasish Halder
- Rare Disease research center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Chanchal Mandal
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
12
|
Cao J, Yan W, Ma X, Huang H, Yan H. Insulin-like Growth Factor 2 mRNA-Binding Protein 2-a Potential Link Between Type 2 Diabetes Mellitus and Cancer. J Clin Endocrinol Metab 2021; 106:2807-2818. [PMID: 34061963 PMCID: PMC8475209 DOI: 10.1210/clinem/dgab391] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Indexed: 12/12/2022]
Abstract
CONTEXT Type 2 diabetes mellitus (T2DM) and cancer share a variety of risk factors and pathophysiological features. It is becoming increasingly accepted that the 2 diseases are related, and that T2DM increases the risk of certain malignancies. OBJECTIVE This review summarizes recent advancements in the elucidation of functions of insulin-like growth factor 2 (IGF-2) messenger RNA (mRNA)-binding protein 2 (IGF2BP2) in T2DM and cancer. METHODS A PubMed review of the literature was conducted, and search terms included IGF2BP2, IMP2, or p62 in combination with cancer or T2DM. Additional sources were identified through manual searches of reference lists. The increased risk of multiple malignancies and cancer-associated mortality in patients with T2DM is believed to be driven by insulin resistance, hyperinsulinemia, hyperglycemia, chronic inflammation, and dysregulation of adipokines and sex hormones. Furthermore, IGF-2 is oncogenic, and its loss-of-function splice variant is protective against T2DM, which highlights the pivotal role of this growth factor in the pathogenesis of these 2 diseases. IGF-2 mRNA-binding proteins, particularly IGF2BP2, are also involved in T2DM and cancer, and single-nucleotide variations (formerly single-nucleotide polymorphisms) of IGF2BP2 are associated with both diseases. Deletion of the IGF2BP2 gene in mice improves their glucose tolerance and insulin sensitivity, and mice with transgenic p62, a splice variant of IGF2BP2, are prone to diet-induced fatty liver disease and hepatocellular carcinoma, suggesting the biological significance of IGF2BP2 in T2DM and cancer. CONCLUSION Accumulating evidence has revealed that IGF2BP2 mediates the pathogenesis of T2DM and cancer by regulating glucose metabolism, insulin sensitivity, and tumorigenesis. This review provides insight into the potential involvement of this RNA binding protein in the link between T2DM and cancer.
Collapse
Affiliation(s)
- Junguo Cao
- Shaanxi Eye Hospital (Xi’an People’s Hospital), Affiliated Guangren Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an 71004, Shaanxi Province, China
- Division of Experimental Neurosurgery, Department of Neurosurgery, University of Heidelberg, Heidelberg 69120, Germany
| | - Weijia Yan
- Shaanxi Eye Hospital (Xi’an People’s Hospital), Affiliated Guangren Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an 71004, Shaanxi Province, China
- Department of Ophthalmology, University of Heidelberg, Heidelberg 69120, Germany
| | - Xiujian Ma
- Division of Molecular Neurogenetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Haiyan Huang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun 130000, China
| | - Hong Yan
- Shaanxi Eye Hospital (Xi’an People’s Hospital), Affiliated Guangren Hospital, School of Medicine, Xi’an Jiaotong University, Xi’an 71004, Shaanxi Province, China
| |
Collapse
|
13
|
Shiek SS, Mani MS, Kabekkodu SP, Dsouza HS. Health repercussions of environmental exposure to lead: Methylation perspective. Toxicology 2021; 461:152927. [PMID: 34492314 DOI: 10.1016/j.tox.2021.152927] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/23/2021] [Accepted: 09/01/2021] [Indexed: 12/15/2022]
Abstract
Lead (Pb) exposure has been a major public health concern for a long time now due to its permanent adverse effects on the human body. The process of lead toxicity has still not been fully understood, but recent advances in Omics technology have enabled researchers to evaluate lead-mediated alterations at the epigenome-wide level. DNA methylation is one of the widely studied and well-understood epigenetic modifications. Pb has demonstrated its ability to induce not just acute deleterious health consequences but also alters the epi-genome such that the disease manifestation happens much later in life as supported by Barkers Hypothesis of the developmental origin of health and diseases. Furthermore, these alterations are passed on to the next generation. Based on previous in-vivo, in-vitro, and human studies, this review provides an insight into the role of Pb in the development of several human disorders.
Collapse
Affiliation(s)
- Sadiya Sadiq Shiek
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Monica Shirley Mani
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Herman S Dsouza
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| |
Collapse
|
14
|
Wang L, Shang C, Pan H, Yang H, Zhu H, Gong F. MicroRNA Expression Profiles in the Subcutaneous Adipose Tissues of Morbidly Obese Chinese Women. Obes Facts 2021; 14:1-15. [PMID: 33550286 PMCID: PMC7983571 DOI: 10.1159/000511772] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/20/2020] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Obesity is a main global health issue and an outstanding cause of morbidity and mortality. Exploring miRNA profiling may help further studies on obesity. METHODS Three morbidly obese and 5 normal-weight Chinese women were enrolled in the microarray testing group. Abdominal subcutaneous adipose tissue (SAT) samples were excised. Total RNAs including miRNAs were extracted. Affymetrix GeneChip miRNA 4.0 Array was used to compare the expression profiles of miRNAs between the 2 groups. Two algorithms, miRanda and TargetScan, were used to predict target messenger RNAs (mRNAs). Bioinformatics analysis was then done based on the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) databases. The sample sizes were further expanded to 8 morbidly obese and 9 normal-weight subjects, and quantitative real-time PCR (qRT-PCR) was utilized to verify the expression of differential miRNAs and target genes. RESULTS As per the microarray assay, 58 miRNAs were differentially expressed in the SAT from the morbidly obese and normal-weight groups (Fold >4, p < 0.01, FDR <0.05); 54 of these were downregulated and 4 were upregulated in morbidly obese subjects. A total of 1,333 target genes were jointly predicted by miRanda and TargetScan. Further bioinformatics analysis showed that the differential miRNAs were involved in 269 significant biological functions and 89 significant signaling pathways. The validation experiment by qRT-PCR showed that the expression levels of miRNA-143-5p, miRNA-143-3p, miRNA-145-5p, and let-7a-5p were downregulated in morbidly obese subjects, consistent with the microarray detection. High-mobility group A2 (HMGA2), a target gene of the downregulated miRNA let-7a-5p, was first found to be upregulated 3.19-fold in the SAT of morbidly obese Chinese women when compared to normal-weight controls. CONCLUSIONS MiRNA downregulation is a hallmark of intact SAT in a morbidly obese state. Transcription (DNA-dependent), small-molecule metabolic processes, the MAPK signaling pathway, and cancer-related pathways may play important roles in the occurrence and development of obesity. For the first time, we proved that HMGA2, a target gene of let-7a-5p, is upregulated in the SAT of morbidly obese Chinese women.
Collapse
Affiliation(s)
- Linjie Wang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Chen Shang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hui Pan
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Hongbo Yang
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Huijuan Zhu
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Fengying Gong
- Key Laboratory of Endocrinology of National Health Commission, Department of Endocrinology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China,
| |
Collapse
|
15
|
Bcl-xL as a Modulator of Senescence and Aging. Int J Mol Sci 2021; 22:ijms22041527. [PMID: 33546395 PMCID: PMC7913597 DOI: 10.3390/ijms22041527] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 01/27/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023] Open
Abstract
Many features of aging result from the incapacity of cells to adapt to stress conditions. When cells are overwhelmed by stress, they can undergo senescence to avoid unrestricted growth of damaged cells. Recent findings have proven that cellular senescence is more than that. A specific grade of senescence promotes embryo development, tissue remodeling and wound healing. However, constant stresses and a weakening immune system can lead to senescence chronicity with aging. The accumulation of senescent cells is directly related to tissue dysfunction and age-related pathologies. Centenarians, the most aged individuals, should accumulate senescent cells and suffer from their deleterious effects, however, they enjoy a compression of morbidity. We have shown that they overexpress B-cell lymphoma-extra large (Bcl-xL). Bcl-xL could avoid an excessive burden of senescent cells through the regulation of intrinsic apoptosis, mitochondrial bioenergetics and oxidative stress. On the other hand, Bcl-xL maintains a fully functional immune system that ensures an efficient clearance of senescent cells. Moreover, there is a paradox, as inhibitors of Bcl-xL have been employed as senolytic agents, which have been shown to protect from aging in animal models. In this review, we aim to discuss how Bcl-xL could modulate senescence-associated harmful effects in centenarians, protecting them from the burden of accumulation of senescent cells.
Collapse
|
16
|
MicroRNA expression patterns in the brown fat of hibernating 13-lined ground squirrels. Genomics 2021; 113:769-781. [PMID: 33529780 DOI: 10.1016/j.ygeno.2021.01.017] [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: 10/05/2020] [Revised: 01/05/2021] [Accepted: 01/24/2021] [Indexed: 11/24/2022]
Abstract
The sequence diversity of microRNAs (miRNAs) allows these potent regulators of mRNA fate to bind multiple transcripts, giving them the power to inhibit diverse cellular processes. Therefore, miRNAs may regulate metabolic rate suppression (also termed torpor), an adaptation used by capable species to reduce energy expenditure, minimize tissue damage, and prolong life. Small RNA-sequencing of brown fat from control (37 °C) and torpid (5-8 °C) ground squirrels revealed a central role for miRNAs in torpor. Unsupervised clustering analysis of all 319 conserved miRNAs showed separation of control and torpor samples, which was supported by PCA analysis. Of the 76 miRNAs that were differentially expressed, 45 were upregulated during torpor. KEGG and GO analyses suggested these miRNAs inhibit genes within the ribosome, oxidative phosphorylation, and glycolysis/gluconeogenesis pathways. Some of the most downregulated miRNAs (miR-1-3p, miR-206 and miR-133a/b) had significant Pearson correlation coefficients, suggesting these myomiRs may be co-expressed in control animals. Only 3 of the 16 enriched KEGG pathways were less targeted by miRNAs during torpor, including cytokine-cytokine receptor interactions and the coagulation and complement cascades, suggesting epigenetic or post-translation modifications may inhibit these potentially damaging processes. Alternatively, their activation could promote damage sensing, wound repair, and improve tissue homeostasis. Overall, miRNA-seq analysis of brown fat revealed a strong role for miRNAs in the downregulation of central metabolic processes necessary for MRS, and highlighted miRNAs that could be inhibited by antagomiRs to promote brown fat activity in potential obesity treatments, or that could be used to replicate torpor in non-hibernating mammals.
Collapse
|
17
|
Mohallem R, Aryal UK. Regulators of TNFα mediated insulin resistance elucidated by quantitative proteomics. Sci Rep 2020; 10:20878. [PMID: 33257747 PMCID: PMC7705713 DOI: 10.1038/s41598-020-77914-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022] Open
Abstract
Obesity is a growing epidemic worldwide and is a major risk factor for several chronic diseases, including diabetes, kidney disease, heart disease, and cancer. Obesity often leads to type 2 diabetes mellitus, via the increased production of proinflammatory cytokines such as tumor necrosis factor-α (TNFα). Our study combines different proteomic techniques to investigate the changes in the global proteome, secretome and phosphoproteome of adipocytes under chronic inflammation condition, as well as fundamental cross-talks between different cellular pathways regulated by chronic TNFα exposure. Our results show that many key regulator proteins of the canonical and non-canonical NF-κB pathways, such as Nfkb2, and its downstream effectors, including Csf-1 and Lgals3bp, directly involved in leukocyte migration and invasion, were significantly upregulated at the intra and extracellular proteomes suggesting the progression of inflammation. Our data provides evidence of several key proteins that play a role in the development of insulin resistance.
Collapse
Affiliation(s)
- Rodrigo Mohallem
- Department of Comparative Pathobiology, Purdue University, West Lafayette, USA
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, USA
| | - Uma K Aryal
- Department of Comparative Pathobiology, Purdue University, West Lafayette, USA.
- Purdue Proteomics Facility, Bindley Bioscience Center, Purdue University, West Lafayette, USA.
| |
Collapse
|
18
|
Qi R, Jiang R, Xiao H, Wang Z, He S, Wang L, Wang Y. Ginsenoside Rg1 protects against d-galactose induced fatty liver disease in a mouse model via FOXO1 transcriptional factor. Life Sci 2020; 254:117776. [PMID: 32437790 DOI: 10.1016/j.lfs.2020.117776] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 02/06/2023]
Abstract
AIMS Rg1 is the most active component of traditional Chinese medicine ginseng, having anti-aging and anti-oxidative stress features in multiple organs. Cellular senescence of hepatocytes is involved in the progression of a wide spectrum of chronic liver diseases. In this study, we investigated the potential benefits and mechanism of action of Rg1 on aging-driven chronic liver diseases. MATERIALS AND METHODS A total of 40 male C57BL/6 mice were randomly divided into four groups: control group; Rg1 group; Rg1+d-gal group; and d-gal group. Blood and liver tissue samples were collected for determination of liver function, biochemical and molecular markers, as well as histopathological investigation. KEY FINDINGS Rg1 played an anti-aging role in reversing d-galactose induced increase in senescence-associated SA-β-gal staining and p53, p21 protein in hepatocytes of mice and sustained mitochondria homeostasis. Meanwhile, Rg1 protected livers from d-galactose caused abnormal elevation of ALT and AST in serum, hepatic steatosis, reduction in hepatic glucose production, hydrogenic degeneration, inflammatory phenomena including senescence-associated secretory phenotype (SASP) IL-1β, IL-6, MCP-1 elevation and lymphocyte infiltration. Furthermore, Rg1 suppressed drastic elevation in FOXO1 phosphorylation resulting in maintaining FOXO1 protein level in the liver after d-galactose treatment, followed by FOXO1 targeted antioxidase SOD and CAT significant up-regulation concurrent with marked decrease in lipid peroxidation marker MDA. SIGNIFICANCE Rg1 exerts pharmaceutic effects of maintaining FOXO1 activity in liver, which enhances anti-oxidation potential of Rg1 to ameliorate SASP and to inhibit inflammation, also promotes metabolic homeostasis, and thus protects livers from senescence induced fatty liver disease. The study provides a potential therapeutic strategy for alleviating chronic liver pathology.
Collapse
Affiliation(s)
- Rongjia Qi
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Medical University, Chongqing 400016, China
| | - Rong Jiang
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Medical University, Chongqing 400016, China
| | - Hanxianzhi Xiao
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Medical University, Chongqing 400016, China
| | - Ziling Wang
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Medical University, Chongqing 400016, China
| | - Siyuan He
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Medical University, Chongqing 400016, China
| | - Lu Wang
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Medical University, Chongqing 400016, China.
| | - Yaping Wang
- Lab of Stem Cell and Tissue Engineering, Department of Histology and Embryology, Medical University, Chongqing 400016, China.
| |
Collapse
|
19
|
Yang Z, Liu X, Wang L, Wang T, Chen Y, Teng X, Li J, Shao L, Hui J, Ye W, Shen Z. The protective effects of HMGA2 in the senescence process of bone marrow-derived mesenchymal stromal cells. J Tissue Eng Regen Med 2020; 14:588-599. [PMID: 32068957 DOI: 10.1002/term.3023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 12/25/2019] [Accepted: 02/03/2020] [Indexed: 12/15/2022]
Abstract
Bone marrow-derived mesenchymal stromal cells (MSCs) have been wildly applied to cell-based strategies for tissue engineering and regenerative medicine; however, they have to undergo the senescence process and thus appeared to be less therapeutic effective. HMGA2, a protein belonged to high mobility group A (HMGA) family, exhibits an inverse expression level related to embryonic development and acts as a developmental regulator in stem cell self-renewal progression. Therefore, we performed senescence-associated β-galactosidase (SA-β-gal) staining, transwell assay, to examine the changes of MSCs in different stages and then over-expressed HMGA2 in MSCs by lentivirus transfection. We found the percentage of SA-β-gal staining positive cells in MSCs from 24-month-old Sprague-Dawley (SD) rats (O-MSCs) was significantly higher compared with MSCs from 2-week-old SD rats (Y-MSCs), and the expression levels of P21 and P53, two senescence-related molecules, were also significantly up-regulated in O-MSCs than in Y-MSCs. In contrast, the HMGA2 expression level in O-MSCs was dramatically down-regulated in contrast to Y-MSCs. In additional, the migration ability in O-MSCs was significantly attenuated than in Y-MSCs. After successfully over-expressed HMGA2 in O-MSCs, the percentage of SA-β-gal staining positive cells and the expression levels of P21 and P53 were reduced, and the migration ability was improved compared with O-MSCs without treatment. Further, mRNA sequencing analysis revealed that overexpression of HMGA2 changed the expression of genes related to cell proliferation and senescence, such as Lyz2, Pf4, Rgs2, and Mstn. Knockdown of Rgs2 in HMGA2 overexpression O-MSCs could antagonize the protective effect of HMGA2 in the senescence process of O-MSCs.
Collapse
Affiliation(s)
- Ziying Yang
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xuan Liu
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Longgang Wang
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Tao Wang
- Department of Cardiology, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Yueqiu Chen
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Xiaomei Teng
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Jingjing Li
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Lianbo Shao
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Jie Hui
- Department of Cardiology, First Affiliated Hospital, Soochow University, Suzhou, China
| | - Wenxue Ye
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| | - Zhenya Shen
- Department of Cardiovascular Surgery, First Affiliated Hospital & Institute for Cardiovascular Science, Soochow University, Suzhou, China
| |
Collapse
|
20
|
Papatheodoridi AM, Chrysavgis L, Koutsilieris M, Chatzigeorgiou A. The Role of Senescence in the Development of Nonalcoholic Fatty Liver Disease and Progression to Nonalcoholic Steatohepatitis. Hepatology 2020; 71:363-374. [PMID: 31230380 DOI: 10.1002/hep.30834] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/17/2019] [Indexed: 12/17/2022]
Abstract
In recent years, cellular senescence has generated a lot of interest among researchers because of its involvement in both the normal aging process and common human diseases. During senescence, cells undergo alterations that include telomere shortening, nuclear area enlargement, and genomic and mitochondrial DNA damage, leading to irreversible cell cycle arrest, and secretion of proinflammatory cytokines. Evidence suggests that the complex process of senescence is involved in the development of a plethora of chronic diseases including metabolic and inflammatory disorders and tumorigenesis. Recently, several human and animal studies have emphasized the involvement of senescence in the pathogenesis and development of liver steatosis including the progression to nonalcoholic steatohepatitis (NASH) as characterized by the additional emergence of inflammation, hepatocyte ballooning, and liver fibrosis. The development of nonalcoholic fatty liver disease (NAFLD) and its progression to NASH are commonly accompanied by several pathophysiological events including metabolic dysregulation and inflammatory phenomena occurring within the liver that may contribute to or derive from cellular senescence, implying that the latter may be both a stimulus and a consequence of the disease. Conclusion: In this review, we summarize the current literature on the impact of cellular senescence in NAFLD/NASH and discuss the effectiveness and safety of novel senolytic drugs and therapeutic options available to delay or treat the disease. Finally, we identify the open questions and issues to be addressed in the near future.
Collapse
Affiliation(s)
| | - Lampros Chrysavgis
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Michael Koutsilieris
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus of TU Dresden, Dresden, Germany
| |
Collapse
|
21
|
Onuzulu CD, Rotimi OA, Rotimi SO. Epigenetic modifications associated with in utero exposure to endocrine disrupting chemicals BPA, DDT and Pb. REVIEWS ON ENVIRONMENTAL HEALTH 2019; 34:309-325. [PMID: 31271561 DOI: 10.1515/reveh-2018-0059] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 04/03/2019] [Indexed: 06/09/2023]
Abstract
Endocrine disrupting chemicals (EDCs) are xenobiotics which adversely modify the hormone system. The endocrine system is most vulnerable to assaults by endocrine disruptors during the prenatal and early development window, and effects may persist into adulthood and across generations. The prenatal stage is a period of vulnerability to environmental chemicals because the epigenome is usually reprogrammed during this period. Bisphenol A (BPA), lead (Pb), and dichlorodiphenyltrichloroethane (DDT) were chosen for critical review because they have become serious public health concerns globally, especially in Africa where they are widely used without any regulation. In this review, we introduce EDCs and describe the various modes of action of EDCs and the importance of the prenatal and developmental windows to EDC exposure. We give a brief overview of epigenetics and describe the various epigenetic mechanisms: DNA methylation, histone modifications and non-coding RNAs, and how each of them affects gene expression. We then summarize findings from previous studies on the effects of prenatal exposure to the endocrine disruptors BPA, Pb and DDT on each of the previously described epigenetic mechanisms. We also discuss how the epigenetic alterations caused by these EDCs may be related to disease processes.
Collapse
Affiliation(s)
- Chinonye Doris Onuzulu
- Department of Biochemistry and Molecular Biology Research Laboratory, Covenant University, Ota, Ogun State, Nigeria
| | - Oluwakemi Anuoluwapo Rotimi
- Department of Biochemistry and Molecular Biology Research Laboratory, Covenant University, Ota, Ogun State, Nigeria
| | - Solomon Oladapo Rotimi
- Department of Biochemistry and Molecular Biology Research Laboratory, Covenant University, Ota, Ogun State, Nigeria
| |
Collapse
|
22
|
Giovos G, Yavropoulou MP, Yovos JG. The role of cellular senescence in diabetes mellitus and osteoporosis: molecular pathways and potential interventions. Hormones (Athens) 2019; 18:339-351. [PMID: 31701490 DOI: 10.1007/s42000-019-00132-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 09/04/2019] [Indexed: 02/06/2023]
Abstract
The improving effectiveness of health care leads inevitably to a rapid increase in the elderly population worldwide. At advanced ages, however, people experience chronic disabilities, which significantly increase the social and economic burden while curtailing survival, independence, and quality of life of the aging population. As aging is a multifactorial process, apart from genetic predisposition, other environmental factors, such as chronic sterile inflammation and cellular senescence, contribute as crucial participants and have been targeted to reverse their deleterious effects on tissue homeostasis and functional integrity. Cellular senescence refers to the essentially irreversible inhibition of cellular proliferation when cells are subjected to extrinsic or endogenous stress. Although the process of cellular senescence has long been known, recent evidence demonstrated that it characterizes many aging phenotypes and that elimination of senescent cells at the tissue level can improve age-related tissue dysfunction. These observations have renewed scientific interest in possible therapeutic interventions. Two major chronic diseases associated with aging that impose an enormous burden on global health systems are type 2 diabetes and osteoporosis. This review presents current data on (i) the underlying molecular mechanisms of cellular senescence, (ii) its relationship to these two endocrine diseases that are today prevalent worldwide, and (iii) future prospects of targeted intervention with the aim of simultaneously improving the progression and prognosis of these serious problems of aging.
Collapse
Affiliation(s)
- Georgios Giovos
- Clinical Research Fellow in Endocrinology, Wisdem Centre, University Hospitals Coventry & Warwickshire, Coventry, UK
| | - Maria P Yavropoulou
- Endocrinology Unit, 1st Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, UOA, LAIKO General Hospital, 17 Agiou Thoma Str., 11527, Athens, Greece.
| | - John G Yovos
- Professor Emeritus in Internal Medicine and Endocrinology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
23
|
Khalid M, Abdollahi M. Epigenetic modifications associated with pathophysiological effects of lead exposure. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2019; 37:235-287. [PMID: 31402779 DOI: 10.1080/10590501.2019.1640581] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Lead (Pb) exposure during different stages of development has demonstrated dose, duration, sex, and tissue-specific pathophysiological outcomes due to altered epigenetic regulation via (a) DNA methylation, (b) histone modifications, (c) miRNAs, and (d) chromatin accessibility. Pb-induced alteration of epigenetic regulation causes neurotoxic and extra-neurotoxic pathophysiological outcomes. Neurotoxic effects of Pb include dysfunction of memory and learning, behavioral disorder, attention deficit hyperactivity disorder, autism spectrum disorder, aging, Alzheimer's disease, tauopathy, and neurodegeneration. Extra-neurotoxic effects of Pb include altered body weight, metabolic disorder, cardiovascular disorders, hematopoietic disorder, and reproductive impairment. Pb exposure either early in life or at any stage of development results in undesirable pathophysiological outcomes that tends to sustain and maintain for a lifetime.
Collapse
Affiliation(s)
- Madiha Khalid
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Li M, Zhao H, Zhao SG, Wei DM, Zhao YR, Huang T, Muhammad T, Yan L, Gao F, Li L, Lu G, Chan WY, Leung PCK, Dunaif A, Liu HB, Chen ZJ. The HMGA2-IMP2 Pathway Promotes Granulosa Cell Proliferation in Polycystic Ovary Syndrome. J Clin Endocrinol Metab 2019; 104:1049-1059. [PMID: 30247605 PMCID: PMC6753588 DOI: 10.1210/jc.2018-00544] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 09/18/2018] [Indexed: 12/19/2022]
Abstract
CONTEXT The high mobility group AT hook 2 (HMGA2) gene was previously identified in a genome-wide association study as a candidate risk gene that might be related to polycystic ovary syndrome (PCOS). Whether HMGA2 contributes to promoting granulosa cell (GC) proliferation in PCOS remains unknown. OBJECTIVE We sought to determine whether HMGA2 is involved in the ovarian dysfunction of PCOS and in the mechanism of increased GC proliferation. PATIENTS AND CELLS mRNA expression was analyzed in ovarian GCs from 96 women with PCOS and 58 healthy controls. Immortalized human GCs (KGN and SVOG cells) were used for the mechanism study. MAIN OUTCOME MEASURES mRNA expression in ovarian GCs was measured using quantitative RT-PCR, and KGN cells were cultured for proliferation assays after overexpression or knockdown of target genes. Protein expression analysis, luciferase assays, and RNA binding protein immunoprecipitation assays were used to confirm the mechanism study. RESULTS HMGA2 and IGF2 mRNA binding protein 2 (IMP2) were highly expressed in the GCs of women with PCOS, and the HMGA2/IMP2 pathway promoted GC proliferation. Cyclin D2 and SERPINE1 mRNA binding protein 1 were regulated by IMP2 and were highly expressed in women with PCOS. CONCLUSIONS The HMGA2/IMP2 pathway was activated in women with PCOS and promoted the proliferation of GCs. This might provide new insights into the dysfunction of GCs in PCOS.
Collapse
Affiliation(s)
- Miao Li
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Han Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Shi-Gang Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Dai-Min Wei
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Yue-Ran Zhao
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Tao Huang
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Tahir Muhammad
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Lei Yan
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
| | - Fei Gao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lei Li
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana
| | - Gang Lu
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Wai-Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
| | - Peter C K Leung
- Department of Obstetrics and Gynaecology, Child and Family Research Institute, University of British Columbia, Vancouver, Canada
| | | | - Hong-Bin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China
- Correspondence and Reprint Requests: Hong-Bin Liu, PhD, or Zi-Jiang Chen, MD, PhD, Center for Reproductive Medicine, Shandong University, No. 157 Jingliu Road, Jinan 250001, China. E-mail: or
| | - Zi-Jiang Chen
- Center for Reproductive Medicine, Shandong University, Jinan, China
- National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, China
- The Key Laboratory of Reproductive Endocrinology (Shandong University), Ministry of Education, Jinan, China
- Correspondence and Reprint Requests: Hong-Bin Liu, PhD, or Zi-Jiang Chen, MD, PhD, Center for Reproductive Medicine, Shandong University, No. 157 Jingliu Road, Jinan 250001, China. E-mail: or
| |
Collapse
|
25
|
Sung Y, Jeong J, Kang RJ, Choi M, Park S, Kwon W, Lee J, Jang S, Park SJ, Kim S, Yi J, Choi S, Lee M, Liu K, Dong Z, Ryoo ZY, Kim MO. Lin28a expression protects against streptozotocin‐induced β‐cell destruction and prevents diabetes in mice. Cell Biochem Funct 2019; 37:139-147. [DOI: 10.1002/cbf.3376] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 12/02/2018] [Accepted: 01/03/2019] [Indexed: 02/06/2023]
Affiliation(s)
- Yonghun Sung
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
| | - Jain Jeong
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
- Core Protein Resources Center, DGIST Daegu Republic of Korea
| | - Ri jin Kang
- Department of Food Science and NutritionKyungpook National University Sangju Republic of Korea
| | - Minjee Choi
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
| | - Song Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
- Core Protein Resources Center, DGIST Daegu Republic of Korea
| | - Wookbong Kwon
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
| | - Jinhee Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
| | - Soyoung Jang
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
| | - Si Jun Park
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
| | - Sung‐Hyun Kim
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
- China‐US (Henan) Hormel Cancer Institute, No.127 Dongming Road Zhengzhou China
| | - Junkoo Yi
- Gyeongsangbukdo Livestock Research Institute Yeongju Republic of Korea
| | | | - Mee‐Hyun Lee
- China‐US (Henan) Hormel Cancer Institute, No.127 Dongming Road Zhengzhou China
| | - Kangdong Liu
- China‐US (Henan) Hormel Cancer Institute, No.127 Dongming Road Zhengzhou China
| | - Zigang Dong
- China‐US (Henan) Hormel Cancer Institute, No.127 Dongming Road Zhengzhou China
| | - Zae Young Ryoo
- School of Life Sciences, BK21 Plus KNU Creative BioResearch GroupKyungpook National University Daegu Republic of Korea
| | - Myoung Ok Kim
- Department of Food Science and NutritionKyungpook National University Sangju Republic of Korea
| |
Collapse
|
26
|
Klee NS, McCarthy CG, Lewis S, McKenzie JL, Vincent JE, Webb RC. Urothelial Senescence in the Pathophysiology of Diabetic Bladder Dysfunction-A Novel Hypothesis. Front Surg 2018; 5:72. [PMID: 30564582 PMCID: PMC6288180 DOI: 10.3389/fsurg.2018.00072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Accepted: 11/12/2018] [Indexed: 12/11/2022] Open
Abstract
Diabetic bladder dysfunction (DBD) is a well-recognized and common symptom affecting up to 50% of all diabetic patients. DBD has a broad range of clinical presentations ranging from overactive to underactive bladder symptoms that develops in middle-aged to elderly patients with long standing and poorly controlled diabetes. Low efficacy of current therapeutics and lifestyle interventions combined with high national healthcare costs highlight the need for more research into bladder dysfunction pathophysiology and novel treatment options. Cellular senescence is an age-related physiologic process in which cells undergo irreversible growth arrest induced by replicative exhaustion and damaging insults. While controlled senescence negatively regulates cell proliferation and promotes tissue regeneration, uncontrolled senescence is known to result in tissue dysfunction through enhanced secretion of inflammatory factors. This review presents previous scientific findings and current hypotheses that characterize diabetic bladder dysfunction. Further, we propose the novel hypothesis that cellular senescence within the urothelial layer of the bladder contributes to the pro-inflammatory/pro-oxidant environment and symptoms of diabetic bladder dysfunction. Our results show increased cellular senescence in the urothelial layer of the bladder; however, whether this phenomenon is the cause or effect of DBD is unknown. The urothelial layer of the bladder is made up of transitional epithelia specialized to contract and expand with demand and plays an active role in transmission by modulating afferent activity. Transition from normal functioning urothelial cells to secretory senescence cells would not only disrupt the barrier function of this layer but may result in altered signaling and sensation of bladder fullness; dysfunction of this layer is known to result in symptoms of frequency and urgency. Future DBD therapeutics may benefit from targeting and preventing early transition of urothelial cells to senescent cells.
Collapse
Affiliation(s)
- Nicole S Klee
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Cameron G McCarthy
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH, United States
| | - Steven Lewis
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Jaine L McKenzie
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States.,Department of Surgery, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - Julie E Vincent
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| | - R Clinton Webb
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, United States
| |
Collapse
|
27
|
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
|
28
|
Wei W, Ji S. Cellular senescence: Molecular mechanisms and pathogenicity. J Cell Physiol 2018; 233:9121-9135. [PMID: 30078211 DOI: 10.1002/jcp.26956] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Cellular senescence is the arrest of normal cell division. Oncogenic genes and oxidative stress, which cause genomic DNA damage and generation of reactive oxygen species, lead to cellular senescence. The senescence-associated secretory phenotype is a distinct feature of senescence. Senescence is normally involved in the embryonic development. Senescent cells can communicate with immune cells to invoke an immune response. Senescence emerges during the aging process in several tissues and organs. In fact, increasing evidence shows that cellular senescence is implicated in aging-related diseases, such as nonalcoholic fatty liver disease, obesity and diabetes, pulmonary hypertension, and tumorigenesis. Cellular senescence can also be induced by microbial infection. During cellular senescence, several signaling pathways, including those of p53, nuclear factor-κB (NF-κB), mammalian target of rapamycin, and transforming growth factor-beta, play important roles. Accumulation of senescent cells can trigger chronic inflammation, which may contribute to the pathological changes in the elderly. Given the variety of deleterious effects caused by cellular senescence in humans, strategies have been proposed to control senescence. In this review, we will focus on recent studies to provide a brief introduction to cellular senescence, including associated signaling pathways and pathology.
Collapse
Affiliation(s)
- Wenqiang Wei
- Laboratory of Cell Signal Transduction, Basic Medical School, Henan University, Kaifeng, Henan, China.,Department of Microbiology, Basic Medical School, Henan University, Kaifeng, Henan, China
| | - Shaoping Ji
- Laboratory of Cell Signal Transduction, Basic Medical School, Henan University, Kaifeng, Henan, China
| |
Collapse
|
29
|
Shakeri H, Lemmens K, Gevaert AB, De Meyer GRY, Segers VFM. Cellular senescence links aging and diabetes in cardiovascular disease. Am J Physiol Heart Circ Physiol 2018; 315:H448-H462. [PMID: 29750567 DOI: 10.1152/ajpheart.00287.2018] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Aging is a powerful independent risk factor for cardiovascular diseases such as atherosclerosis and heart failure. Concomitant diabetes mellitus strongly reinforces this effect of aging on cardiovascular disease. Cellular senescence is a fundamental mechanism of aging and appears to play a crucial role in the onset and prognosis of cardiovascular disease in the context of both aging and diabetes. Senescent cells are in a state of cell cycle arrest but remain metabolically active by secreting inflammatory factors. This senescence-associated secretory phenotype is a trigger of chronic inflammation, oxidative stress, and decreased nitric oxide bioavailability. A complex interplay between these three mechanisms results in age- and diabetes-associated cardiovascular damage. In this review, we summarize current knowledge on cellular senescence and its secretory phenotype, which might be the missing link between aging and diabetes contributing to cardiovascular disease.
Collapse
Affiliation(s)
- Hadis Shakeri
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium
| | - Katrien Lemmens
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium
| | - Andreas B Gevaert
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium.,Laboratory for Cellular and Molecular Cardiology, Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium
| | - Vincent F M Segers
- Laboratory of Physiopharmacology, University of Antwerp , Antwerp , Belgium.,Department of Cardiology, University Hospital Antwerp, Edegem, Belgium
| |
Collapse
|
30
|
Zeng CP, Chen YC, Lin X, Greenbaum J, Chen YP, Peng C, Wang XF, Zhou R, Deng WM, Shen J, Deng HW. Increased identification of novel variants in type 2 diabetes, birth weight and their pleiotropic loci. J Diabetes 2017; 9:898-907. [PMID: 27896934 PMCID: PMC5841537 DOI: 10.1111/1753-0407.12510] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 11/12/2016] [Accepted: 11/24/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Clinical and epidemiological findings point to an association between type 2 diabetes (T2D) and low birth weight. However, the nature of the relationship is largely unknown. The aim of this study was to identify novel single nucleotide polymorphisms (SNPs) in T2D and birth weight, and their pleiotropic loci. METHODS A pleiotropy-informed conditional false discovery rate (cFDR) method was applied to two independent genome-wide association studies (GWAS) summary statistics of T2D (n = 149 821) and birth weight (n = 26 836). RESULTS A conditional Q-Q plot showed strong enrichment of genetic variants in T2D conditioned on different levels of association with birth weight. 133 T2D-associated SNPs, including 120 novel SNPs, were identified with a significance threshold of cFDR < 0.05; 13 significant birth weight-associated SNPs, including 12 novel SNPs (cFDR < 0.05) were identified. Conjunctional cFDR (ccFDR) analysis identified nine pleiotropic loci, including seven novel loci, shared by both T2D and birth weight (ccFDR < 0.05). Two novel SNPs located at the CDK5 regulatory subunit-associated protein 1-like 1 (CDKAL1; rs1012635; cFDR < 0.05) and adenylate cyclase 5 (ADCY5; rs4677887; cFDR < 0.05) genes are of note. These two genes increase the risk of T2D and low birth weight through the pathway of the "fetal insulin hypothesis." CONCLUSION Several pleiotropic loci were identified between T2D and birth weight by leveraging GWAS results. The results make it possible to explain a greater proportion of trait heritability and improve our understanding of the shared pathophysiology between T2D and birth weight.
Collapse
Affiliation(s)
- Chun-Ping Zeng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Endocrinology and Metabolism, Affiliated Nanhai Hospital of Southern Medical University, Guangzhou, China
| | - Yuan-Cheng Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xu Lin
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Jonathan Greenbaum
- Department of Biostatistics and Bioinformatics, Center for Bioinformatics and Genomics, Tulane University, New Orleans, Louisiana, USA
| | - You-Ping Chen
- Department of Endocrinology and Metabolism, Affiliated Nanhai Hospital of Southern Medical University, Guangzhou, China
| | - Cheng Peng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Xia-Fang Wang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Rou Zhou
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Wei-Min Deng
- Department of Rehabilitation, General Hospital of Guangzhou Military Command of Chinese PLA, Guangzhou, China
| | - Jie Shen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Hong-Wen Deng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
- Department of Biostatistics and Bioinformatics, Center for Bioinformatics and Genomics, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
31
|
Montrose L, Faulk C, Francis J, Dolinoy D. Perinatal lead (Pb) exposure results in sex and tissue-dependent adult DNA methylation alterations in murine IAP transposons. ENVIRONMENTAL AND MOLECULAR MUTAGENESIS 2017; 58:540-550. [PMID: 28833526 PMCID: PMC5784428 DOI: 10.1002/em.22119] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 05/25/2017] [Accepted: 05/25/2017] [Indexed: 05/17/2023]
Abstract
Epidemiological and animal data suggest that adult chronic disease is influenced by early-life exposure-induced changes to the epigenome. Previously, we observed that perinatal lead (Pb) exposure results in persistent murine metabolic- and activity-related effects. Using phylogenetic and DNA methylation analysis, we have also identified novel intracisternal A particle (IAP) retrotransposons exhibiting regions of variable methylation as candidate loci for environmental effects on the epigenome. Here, we now evaluate brain and kidney DNA methylation profiles of four representative IAPs in adult mice exposed to human physiologically relevant levels of Pb two weeks prior to mating through lactation. When IAPs across the genome were evaluated globally, average (sd) methylation levels were 92.84% (3.74) differing by tissue (P < 0.001), but not sex or dose. By contrast, the four individual IAPs displayed tissue-specific Pb and sex effects. Medium Pb-exposed mice had 3.86% less brain methylation at IAP 110 (P < 0.01), while high Pb-exposed mice had 2.83% less brain methylation at IAP 236 (P = 0.01) and 1.77% less at IAP 506 (P = 0.05). Individual IAP DNA methylation differed by sex for IAP 110 in the brain and kidney, IAP 236 in the kidney, and IAP 1259 in the kidney. Using Tomtom, we identified three binding motifs that matched to each of our novel IAPs impacted by Pb, one of which (HMGA2) has been linked to metabolic-related conditions in both mice and humans. Thus, these recently identified IAPs display tissue-specific environmental lability as well as sex-specific differences supporting an epigenetic link between early exposure to Pb and later-in-life health outcomes. Environ. Mol. Mutagen. 58:540-550, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- L. Montrose
- Environmental Health Sciences, University of Michigan
| | - C. Faulk
- Animal Science, University of Minnesota
| | - J. Francis
- Environmental Health Sciences, University of Michigan
| | - D.C. Dolinoy
- Environmental Health Sciences, University of Michigan
- Nutritional Sciences, University of Michigan
- Corresponding author: Dana C. Dolinoy, 1415 Washington Heights, Ann Arbor, Michigan 48109-2029, Tel: 734 647-3155,
| |
Collapse
|
32
|
Childs BG, Gluscevic M, Baker DJ, Laberge RM, Marquess D, Dananberg J, van Deursen JM. Senescent cells: an emerging target for diseases of ageing. Nat Rev Drug Discov 2017; 16:718-735. [PMID: 28729727 PMCID: PMC5942225 DOI: 10.1038/nrd.2017.116] [Citation(s) in RCA: 764] [Impact Index Per Article: 109.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Chronological age represents the single greatest risk factor for human disease. One plausible explanation for this correlation is that mechanisms that drive ageing might also promote age-related diseases. Cellular senescence, which is a permanent state of cell cycle arrest induced by cellular stress, has recently emerged as a fundamental ageing mechanism that also contributes to diseases of late life, including cancer, atherosclerosis and osteoarthritis. Therapeutic strategies that safely interfere with the detrimental effects of cellular senescence, such as the selective elimination of senescent cells (SNCs) or the disruption of the SNC secretome, are gaining significant attention, with several programmes now nearing human clinical studies.
Collapse
Affiliation(s)
| | | | - Darren J Baker
- Departments of Biochemistry and Molecular Biology, Mayo Clinic
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st St. SW, Rochester, Minnesota 55905, USA
| | - Remi-Martin Laberge
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Dan Marquess
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Jamie Dananberg
- Unity Biotechnology, 3280 Bayshore Boulevard Suite 100, Brisbane, California 94005, USA
| | - Jan M van Deursen
- Departments of Biochemistry and Molecular Biology, Mayo Clinic
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, 200 1st St. SW, Rochester, Minnesota 55905, USA
| |
Collapse
|
33
|
Zhu Y, Xiong K, Shi J, Cui Q, Xue L. A potential role of microRNAs in protein accumulation in cellular senescence analyzed by bioinformatics. PLoS One 2017; 12:e0179034. [PMID: 28591170 PMCID: PMC5462445 DOI: 10.1371/journal.pone.0179034] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/23/2017] [Indexed: 12/25/2022] Open
Abstract
Cellular senescence is an important protective mechanism against cell proliferation and has critical roles in aging and aging-related disease. Recently, one interesting observation is that the protein abundance is higher in senescent cells than that in young cells. So far, some factors were presented to interpret this observation, such as active protein synthesis linked with autophagy, mTOR, and oxidative stress. Here, applying bioinformatic analysis of microRNA profiles in young cells and aging cells, we revealed that globally senescent cells show lower miRNA abundance than that in young cells, suggesting that the repression of protein synthesis by miRNA in senescent cells could be largely attenuated. This finding provides clues that protein accumulation in cellular senescence could be associated with lower miRNA abundance in aging cells.
Collapse
Affiliation(s)
- Yuequan Zhu
- Medical Research Center, Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Kai Xiong
- Medical Research Center, Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
| | - Jiangcheng Shi
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Qinghua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, China
- * E-mail: (LX); (QC)
| | - Lixiang Xue
- Medical Research Center, Department of Radiation Oncology, Peking University Third Hospital, Beijing, China
- * E-mail: (LX); (QC)
| |
Collapse
|
34
|
Sturmlechner I, Durik M, Sieben CJ, Baker DJ, van Deursen JM. Cellular senescence in renal ageing and disease. Nat Rev Nephrol 2016; 13:77-89. [DOI: 10.1038/nrneph.2016.183] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
35
|
Xi Y, Shen W, Ma L, Zhao M, Zheng J, Bu S, Hino S, Nakao M. HMGA2 promotes adipogenesis by activating C/EBPβ-mediated expression of PPARγ. Biochem Biophys Res Commun 2016; 472:617-23. [PMID: 26966068 DOI: 10.1016/j.bbrc.2016.03.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Accepted: 03/06/2016] [Indexed: 12/11/2022]
Abstract
Adipogenesis is orchestrated by a highly ordered network of transcription factors including peroxisome-proliferator activated receptor-gamma (PPARγ) and CCAAT-enhancer binding protein (C/EBP) family proteins. High mobility group protein AT-hook 2 (HMGA2), an architectural transcription factor, has been reported to play an essential role in preadipocyte proliferation, and its overexpression has been implicated in obesity in mice and humans. However, the direct role of HMGA2 in regulating the gene expression program during adipogenesis is not known. Here, we demonstrate that HMGA2 is required for C/EBPβ-mediated expression of PPARγ, and thus promotes adipogenic differentiation. We observed a transient but marked increase of Hmga2 transcript at an early phase of differentiation of mouse 3T3-L1 preadipocytes. Importantly, Hmga2 knockdown greatly impaired adipocyte formation, while its overexpression promoted the formation of mature adipocytes. We found that HMGA2 colocalized with C/EBPβ in the nucleus and was required for the recruitment of C/EBPβ to its binding element at the Pparγ2 promoter. Accordingly, HMGA2 and C/EBPβ cooperatively enhanced the Pparγ2 promoter activity. Our results indicate that HMGA2 is an essential constituent of the adipogenic transcription factor network, and thus its function may be affected during the course of obesity.
Collapse
Affiliation(s)
- Yang Xi
- Diabetes Center, and Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Wanjing Shen
- Diabetes Center, and Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Lili Ma
- Diabetes Center, and Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Ming Zhao
- Diabetes Center, and Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Jiachen Zheng
- Diabetes Center, and Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo 315211, China
| | - Shizhong Bu
- Diabetes Center, and Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo 315211, China.
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811, Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, 860-0811, Japan; Core Research for Evolutional Science and Technology (CREST), Japan Agency for Medical Research and Development, Tokyo, Japan.
| |
Collapse
|
36
|
Patel RS, Carter G, El Bassit G, Patel AA, Cooper DR, Murr M, Patel NA. Adipose-derived stem cells from lean and obese humans show depot specific differences in their stem cell markers, exosome contents and senescence: role of protein kinase C delta (PKCδ) in adipose stem cell niche. Stem Cell Investig 2016; 3:2. [PMID: 27358894 DOI: 10.3978/j.issn.2306-9759.2016.01.02] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 01/21/2016] [Indexed: 01/23/2023]
Abstract
BACKGROUND Adipose-derived stem cells (ASC) and its exosomes are gaining utmost importance in the field of regenerative medicine. The ASCs tested for their potential in wound healing are predominantly derived from the subcutaneous depot of lean donors. However, it is important to characterize the ASC derived from different adipose depots as these depots have clinically distinct roles. METHODS We characterized the ASC derived from subcutaneous and omental depots from a lean donor (sc-ASCn and om-ASCn) and compared it to the ASC derived from an obese donor (sc-ASCo and om-ASCo) using flow cytometry and real time qPCR. RESULTS We show that stem cell markers Oct4, Sal4, Sox15, KLF4 and BMI1 have distinct expression patterns in each ASC. We evaluated the secretome of the ASC and characterized their secreted exosomes. We show long noncoding RNAs (lncRNAs) are secreted by ASC and their expression varied between the ASC's derived from different depots. Protein kinase C delta (PKCδ) regulates the mitogenic signals in stem cells. We evaluated the effect of silencing PKCδ in sc-ASCn, om-ASCn, sc-ASCo and om-ASCo. Using β-galactosidase staining, we evaluated the percentage of senescent cells in sc-ASCn, om-ASCn, sc-ASCo and om-ASCo. Our results also indicated that silencing PKCδ increases the percentage of senescent cells. CONCLUSIONS Our case-specific study demonstrates a role of PKCδ in maintaining the adipose stem cell niche and importantly demonstrates depot-specific differences in adipose stem cells and their exosome content.
Collapse
Affiliation(s)
- Rekha S Patel
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Gay Carter
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Ghattas El Bassit
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Achintya A Patel
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Denise R Cooper
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Michel Murr
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| | - Niketa A Patel
- 1 Department of Molecular Medicine, University of South Florida, Tampa, FL, USA ; 2 James A. Haley Veterans Hospital, Tampa, FL, USA ; 3 Honors College, University of South Florida, Tampa, FL, USA ; 4 Department of Surgery, University of South Florida, Tampa, FL, USA
| |
Collapse
|
37
|
Adipose tissue hypoxia and low-grade inflammation: a possible mechanism for ethanol-related glucose intolerance? Br J Nutr 2015; 113:1355-64. [PMID: 25989996 DOI: 10.1017/s000711451500077x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The exact mechanism of ethanol's effects on glucose tolerance has not been well determined. The present study focuses for the first time on hypoxia and low-grade inflammation in adipose tissue (AT). In the in vivo experiments, twenty-four male Wistar rats were randomly allocated into control and ethanol feeding groups. Ethanol-treated rats received edible ethanol once a day at a total dosage of 5 g/kg per d, and the controls received distilled water. Ethanol volumes were adjusted every week. At the end of 8 weeks, we carried out an oral glucose tolerance test. Blood and AT were collected for measuring hypoxia-inducible factor-1α (HIF-1α), GLUT1, TNF-α, IL-6, leptin and vascular endothelial growth factor (VEGF). In the in vitro experiments, differentiated OP9 adipocytes were incubated with 100 mm of ethanol for 48 h; the media and cells were then collected for measuring HIF-1α, GLUT1, TNF-α and IL-6. The results showed that long-term ethanol consumption impaired glucose tolerance in rats. Ethanol consumption had little influence on body weight, but both epididymal and perirenal AT were markedly enlarged in the ethanol-treated rats as compared to the controls. Visceral adipose tissue (VAT) had accumulated, and the protein levels of HIF-1α and GLUT1, the indicators of hypoxia in rat epididymal AT and OP9 adipocytes, were elevated. Secondary to the AT hypoxia, the levels of inflammation-related adipokines, such as TNF-α, IL-6, leptin and VEGF, were increased. Based on these findings, we conclude that VAT hypoxia and low-grade inflammation might be a new mechanism in the treatment of ethanol-related diabetes.
Collapse
|
38
|
Senescent cells: SASPected drivers of age-related pathologies. Biogerontology 2014; 15:627-42. [PMID: 25217383 DOI: 10.1007/s10522-014-9529-9] [Citation(s) in RCA: 140] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 09/01/2014] [Indexed: 12/19/2022]
Abstract
The progression of physiological ageing is driven by intracellular aberrations including telomere attrition, genomic instability, epigenetic alterations and loss of proteostasis. These in turn damage cells and compromise their functionality. Cellular senescence, a stable irreversible cell-cycle arrest, is elicited in damaged cells and prevents their propagation in the organism. Under normal conditions, senescent cells recruit the immune system which facilitates their removal from tissues. Nevertheless, during ageing, tissue-residing senescent cells tend to accumulate, and might negatively impact their microenvironment via profound secretory phenotype with pro-inflammatory characteristics, termed senescence-associated secretory phenotype (SASP). Indeed, senescent cells are mostly abundant at sites of age-related pathologies, including degenerative disorders and malignancies. Interestingly, studies on progeroid mice indicate that selective elimination of senescent cells can delay age-related deterioration. This suggests that chronic inflammation induced by senescent cells might be a main driver of these pathologies. Importantly, senescent cells accumulate as a result of deficient immune surveillance, and their removal is increased upon the use of immune stimulatory agents. Insights into mechanisms of senescence surveillance could be combined with current approaches for cancer immunotherapy to propose new preventive and therapeutic strategies for age-related diseases.
Collapse
|
39
|
Abstract
Recent discoveries are redefining our view of cellular senescence as a trigger of tissue remodelling that acts during normal embryonic development and upon tissue damage. To achieve this, senescent cells arrest their own proliferation, recruit phagocytic immune cells and promote tissue renewal. This sequence of events - senescence, followed by clearance and then regeneration - may not be efficiently completed in aged tissues or in pathological contexts, thereby resulting in the accumulation of senescent cells. Increasing evidence indicates that both pro-senescent therapies and antisenescent therapies can be beneficial. In cancer and during active tissue repair, pro-senescent therapies contribute to minimize the damage by limiting proliferation and fibrosis, respectively. Conversely, antisenescent therapies may help to eliminate accumulated senescent cells and to recover tissue function.
Collapse
|
40
|
Schleinitz D, Böttcher Y, Blüher M, Kovacs P. The genetics of fat distribution. Diabetologia 2014; 57:1276-86. [PMID: 24632736 DOI: 10.1007/s00125-014-3214-z] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Accepted: 02/18/2014] [Indexed: 12/22/2022]
Abstract
Fat stored in visceral depots makes obese individuals more prone to complications than subcutaneous fat. There is good evidence that body fat distribution (FD) is controlled by genetic factors. WHR, a surrogate measure of FD, shows significant heritability of up to ∼60%, even after adjusting for BMI. Genetic variants have been linked to various forms of altered FD such as lipodystrophies; however, the polygenic background of visceral obesity has only been sparsely investigated in the past. Recent genome-wide association studies (GWAS) for measures of FD revealed numerous loci harbouring genes potentially regulating FD. In addition, genes with fat depot-specific expression patterns (in particular subcutaneous vs visceral adipose tissue) provide plausible candidate genes involved in the regulation of FD. Many of these genes are differentially expressed in various fat compartments and correlate with obesity-related traits, thus further supporting their role as potential mediators of metabolic alterations associated with a distinct FD. Finally, developmental genes may at a very early stage determine specific FD in later life. Indeed, genes such as TBX15 not only manifest differential expression in various fat depots, but also correlate with obesity and related traits. Moreover, recent GWAS identified several polymorphisms in developmental genes (including TBX15, HOXC13, RSPO3 and CPEB4) strongly associated with FD. More accurate methods, including cardiometabolic imaging, for assessment of FD are needed to promote our understanding in this field, where the main focus is now to unravel the yet unknown biological function of these novel 'fat distribution genes'.
Collapse
Affiliation(s)
- Dorit Schleinitz
- Integrated Research and Treatment Center (IFB) AdiposityDiseases, University of Leipzig, Liebigstr. 21, 04103, Leipzig, Germany
| | | | | | | |
Collapse
|
41
|
Zhu Y, Armstrong JL, Tchkonia T, Kirkland JL. Cellular senescence and the senescent secretory phenotype in age-related chronic diseases. Curr Opin Clin Nutr Metab Care 2014; 17:324-8. [PMID: 24848532 DOI: 10.1097/mco.0000000000000065] [Citation(s) in RCA: 192] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Possible mechanisms in cellular senescence and the senescence-associated secretory phenotype (SASP) that drive and promote chronic inflammation in multiple age-related chronic diseases are considered. RECENT FINDINGS A series of studies about the SASP indicate that senescent cells may be involved in the development of chronic inflammatory diseases associated with aging. SUMMARY Aging is a complex biological process accompanied by a state of chronic, low-grade, 'sterile' inflammation, which is a major contributor to the development of many age-related chronic disorders including atherosclerosis, osteoarthritis, Alzheimer's disease, type 2 diabetes, cancers, and others. It appears that cellular senescence plays a role in causing inflammation through the SASP. A better understanding of the contribution of senescent cells to the pathologies of chronic inflammatory disorders could have potentially profound diagnostic and therapeutic implications.
Collapse
Affiliation(s)
- Yi Zhu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | | | | | | |
Collapse
|