1
|
Fatema K, Haidar Z, Tanim MTH, Nath SD, Sajib AA. Unveiling the link between arsenic toxicity and diabetes: an in silico exploration into the role of transcription factors. Toxicol Res 2024; 40:653-672. [PMID: 39345741 PMCID: PMC11436564 DOI: 10.1007/s43188-024-00255-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 04/10/2024] [Accepted: 07/10/2024] [Indexed: 10/01/2024] Open
Abstract
Arsenic-induced diabetes, despite being a relatively newer finding, is now a growing area of interest, owing to its multifaceted nature of development and the diversity of metabolic conditions that result from it, on top of the already complicated manifestation of arsenic toxicity. Identification and characterization of the common and differentially affected cellular metabolic pathways and their regulatory components among various arsenic and diabetes-associated complications may aid in understanding the core molecular mechanism of arsenic-induced diabetes. This study, therefore, explores the effects of arsenic on human cell lines through 14 transcriptomic datasets containing 160 individual samples using in silico tools to take a systematic, deeper look into the pathways and genes that are being altered. Among these, we especially focused on the role of transcription factors due to their diverse and multifaceted roles in biological processes, aiming to comprehensively investigate the underlying mechanism of arsenic-induced diabetes as well as associated health risks. We present a potential mechanism heavily implying the involvement of the TGF-β/SMAD3 signaling pathway leading to cell cycle alterations and the NF-κB/TNF-α, MAPK, and Ca2+ signaling pathways underlying the pathogenesis of arsenic-induced diabetes. This study also presents novel findings by suggesting potential associations of four transcription factors (NCOA3, PHF20, TFDP1, and TFDP2) with both arsenic toxicity and diabetes; five transcription factors (E2F5, ETS2, EGR1, JDP2, and TFE3) with arsenic toxicity; and one transcription factor (GATA2) with diabetes. The novel association of the transcription factors and proposed mechanism in this study may serve as a take-off point for more experimental evidence needed to understand the in vivo cellular-level diabetogenic effects of arsenic. Supplementary Information The online version contains supplementary material available at 10.1007/s43188-024-00255-y.
Collapse
Affiliation(s)
- Kaniz Fatema
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Zinia Haidar
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Md Tamzid Hossain Tanim
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Sudipta Deb Nath
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| | - Abu Ashfaqur Sajib
- Department of Genetic Engineering & Biotechnology, University of Dhaka, Dhaka, 1000 Bangladesh
| |
Collapse
|
2
|
Howes AM, Dea NC, Ghosh D, Krishna K, Wang Y, Li Y, Morrison B, Toussaint KC, Dawson MR. Fibroblast senescence-associated extracellular matrix promotes heterogeneous lung niche. APL Bioeng 2024; 8:026119. [PMID: 38855444 PMCID: PMC11161856 DOI: 10.1063/5.0204393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/24/2024] [Indexed: 06/11/2024] Open
Abstract
Senescent cell accumulation in the pulmonary niche is associated with heightened susceptibility to age-related disease, tissue alterations, and ultimately a decline in lung function. Our current knowledge of senescent cell-extracellular matrix (ECM) dynamics is limited, and our understanding of how senescent cells influence spatial ECM architecture changes over time is incomplete. Herein is the design of an in vitro model of senescence-associated extracellular matrix (SA-ECM) remodeling using a senescent lung fibroblast-derived matrix that captures the spatiotemporal dynamics of an evolving senescent ECM architecture. Multiphoton second-harmonic generation microscopy was utilized to examine the spatial and temporal dynamics of fibroblast SA-ECM remodeling, which revealed a biphasic process that established a disordered and heterogeneous architecture. Additionally, we observed that inhibition of transforming growth factor-β signaling during SA-ECM remodeling led to improved local collagen fiber organization. Finally, we examined patient samples diagnosed with pulmonary fibrosis to further tie our results of the in vitro model to clinical outcomes. Moreover, we observed that the senescence marker p16 is correlated with local collagen fiber disorder. By elucidating the temporal dynamics of SA-ECM remodeling, we provide further insight on the role of senescent cells and their contributions to pathological ECM remodeling.
Collapse
Affiliation(s)
| | - Nova C. Dea
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Deepraj Ghosh
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Krishangi Krishna
- School of Engineering, Brown University, Providence, Rhode Island 02912, USA
| | - Yihong Wang
- Department of Pathology and Laboratory Medicine, Brown University, Providence, Rhode Island 02912, USA
| | - Yanxi Li
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Braxton Morrison
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| | - Kimani C. Toussaint
- School of Engineering, Brown University, Providence, Rhode Island 02912, USA
| | - Michelle R. Dawson
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island 029012, USA
| |
Collapse
|
3
|
Liu B, Xie D, Huang X, Jin S, Dai Y, Sun X, Li D, Bennett AM, Diano S, Huang Y. Skeletal muscle TET3 promotes insulin resistance through destabilisation of PGC-1α. Diabetologia 2024; 67:724-737. [PMID: 38216792 PMCID: PMC10904493 DOI: 10.1007/s00125-023-06073-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/17/2023] [Indexed: 01/14/2024]
Abstract
AIM/HYPOTHESIS The peroxisome proliferator-activated receptor-γ coactivator α (PGC-1α) plays a critical role in the maintenance of glucose, lipid and energy homeostasis by orchestrating metabolic programs in multiple tissues in response to environmental cues. In skeletal muscles, PGC-1α dysregulation has been associated with insulin resistance and type 2 diabetes but the underlying mechanisms have remained elusive. This research aims to understand the role of TET3, a member of the ten-eleven translocation (TET) family dioxygenases, in PGC-1α dysregulation in skeletal muscles in obesity and diabetes. METHODS TET expression levels in skeletal muscles were analysed in humans with or without type 2 diabetes, as well as in mouse models of high-fat diet (HFD)-induced or genetically induced (ob/ob) obesity/diabetes. Muscle-specific Tet3 knockout (mKD) mice were generated to study TET3's role in muscle insulin sensitivity. Genome-wide expression profiling (RNA-seq) of muscle tissues from wild-type (WT) and mKD mice was performed to mine deeper insights into TET3-mediated regulation of muscle insulin sensitivity. The correlation between PGC-1α and TET3 expression levels was investigated using muscle tissues and in vitro-derived myotubes. PGC-1α phosphorylation and degradation were analysed using in vitro assays. RESULTS TET3 expression was elevated in skeletal muscles of humans with type 2 diabetes and in HFD-fed and ob/ob mice compared with healthy controls. mKD mice exhibited enhanced glucose tolerance, insulin sensitivity and resilience to HFD-induced insulin resistance. Pathway analysis of RNA-seq identified 'Mitochondrial Function' and 'PPARα Pathway' to be among the top biological processes regulated by TET3. We observed higher PGC-1α levels (~25%) in muscles of mKD mice vs WT mice, and lower PGC-1α protein levels (~25-60%) in HFD-fed or ob/ob mice compared with their control counterparts. In human and murine myotubes, increased PGC-1α levels following TET3 knockdown contributed to improved mitochondrial respiration and insulin sensitivity. TET3 formed a complex with PGC-1α and interfered with its phosphorylation, leading to its destabilisation. CONCLUSIONS/INTERPRETATION Our results demonstrate an essential role for TET3 in the regulation of skeletal muscle insulin sensitivity and suggest that TET3 may be used as a potential therapeutic target for the metabolic syndrome. DATA AVAILABILITY Sequences are available from the Gene Expression Omnibus ( https://www.ncbi.nlm.nih.gov/geo/ ) with accession number of GSE224042.
Collapse
Affiliation(s)
- Beibei Liu
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Center of Reproductive Medicine, National Health Commission Key Laboratory of Advanced Reproductive Medicine and Fertility, Shengjing Hospital of China Medical University, Shenyang, China
| | - Di Xie
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Department of Reproductive Medicine, General Hospital of Central Theater Command, Wuhan, Hubei, China
| | - Xinmei Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Department of Endocrinology, Fifth People's Hospital of Shanghai, Fudan University School of Medicine, Shanghai, China
| | - Sungho Jin
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Yangyang Dai
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xiaoli Sun
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Center of Reproductive Medicine, Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, Jiangsu, China
| | - Da Li
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA
- Center of Reproductive Medicine, National Health Commission Key Laboratory of Advanced Reproductive Medicine and Fertility, Shengjing Hospital of China Medical University, Shenyang, China
| | - Anton M Bennett
- Departments of Pharmacology and of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA
| | - Sabrina Diano
- Institute of Human Nutrition, Columbia University Irving Medical Center, New York, NY, USA
| | - Yingqun Huang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
- Yale Center for Molecular and Systems Metabolism, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Das SS, Das SK. Common and ethnic-specific derangements in skeletal muscle transcriptome associated with obesity. Int J Obes (Lond) 2024; 48:330-338. [PMID: 37993634 DOI: 10.1038/s41366-023-01417-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 10/25/2023] [Accepted: 11/07/2023] [Indexed: 11/24/2023]
Abstract
BACKGROUND Obesity is a common disease with a higher prevalence among African Americans. Obesity alters cellular function in many tissues, including skeletal muscle, and is a risk factor for many life-threatening diseases, including cardiovascular disease and diabetes. The similarities and differences in molecular mechanisms that may explain ethnic disparities in obesity between African and European ancestry individuals have not been studied. METHODS In this study, data from transcriptome-wide analyses on skeletal muscle tissues from well-powered human cohorts were used to compare genes and biological pathways affected by obesity in European and African ancestry populations. Data on obesity-induced differentially expressed transcripts and GWAS-identified SNPs were integrated to prioritize target genes for obesity-associated genetic variants. RESULTS Linear regression analysis in the FUSION (European, N = 301) and AAGMEx (African American, N = 256) cohorts identified a total of 2569 body mass index (BMI)-associated transcripts (q < 0.05), of which 970 genes (at p < 0.05) are associated in both cohorts, and the majority showed the same direction of effect on BMI. Biological pathway analyses, including over-representation and gene-set enrichment analyses, identified enrichment of protein synthesis pathways (e.g., ribosomal function) and the ceramide signaling pathway in both cohorts among BMI-associated down- and up-regulated transcripts, respectively. A comparison using the IPA-tool suggested the activation of inflammation pathways only in Europeans with obesity. Interestingly, these analyses suggested repression of the mitochondrial oxidative phosphorylation pathway in Europeans but showed its activation in African Americans. Integration of SNP-to-Gene analyses-predicted target genes for obesity-associated genetic variants (GWAS-identified SNPs) and BMI-associated transcripts suggested that these SNPs might cause obesity by altering the expression of 316 critical target genes (e.g., GRB14) in the muscle. CONCLUSIONS This study provides a replication of obesity-associated transcripts and biological pathways in skeletal muscle across ethnicities, but also identifies obesity-associated processes unique in either African or European ancestry populations.
Collapse
Affiliation(s)
- Sreejon S Das
- The School of Biotechnology at Atkins, Atkins Academic and Technology High, Winston-Salem, NC, 27101, USA
| | - Swapan K Das
- Department of Internal Medicine, Section of Endocrinology and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA.
| |
Collapse
|
5
|
Sharma K, Saini N, Hasija Y. Identifying the mitochondrial metabolism network by integration of machine learning and explainable artificial intelligence in skeletal muscle in type 2 diabetes. Mitochondrion 2024; 74:101821. [PMID: 38040172 DOI: 10.1016/j.mito.2023.11.004] [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: 06/26/2023] [Revised: 10/04/2023] [Accepted: 11/26/2023] [Indexed: 12/03/2023]
Abstract
Imbalance in glucose metabolism and insulin resistance are two primary features of type 2 diabetes/diabetes mellitus. Its etiology is linked to mitochondrial dysfunction in skeletal muscle tissue. The mitochondria are vital organelles involved in ATP synthesis and metabolism. The underlying biological pathways leading to mitochondrial dysfunction in type 2 diabetes can help us understand the pathophysiology of the disease. In this study, the mitochondrial gene expression dataset were retrieved from the GSE22309, GSE25462, and GSE18732 using Mitocarta 3.0, focusing specifically on genes that are associated with mitochondrial function in type 2 disease. Feature selection on the expression dataset of skeletal muscle tissue from 107 control patients and 70 type 2 diabetes patients using the XGBoost algorithm having the highest accuracy. For interpretation and analysis of results linked to the disease by examining the feature importance deduced from the model was done using SHAP (SHapley Additive exPlanations). Next, to comprehend the biological connections, study of protein-protien and mRNA-miRNA networks was conducted using String and Mienturnet respectively. The analysis revealed BDH1, YARS2, AKAP10, RARS2, MRPS31, were potential mitochondrial target genes among the other twenty genes. These genes are mainly involved in the transport and organization of mitochondria, regulation of its membrane potential, and intrinsic apoptotic signaling etc. mRNA-miRNA interaction network revealed a significant role of miR-375; miR-30a-5p; miR-16-5p; miR-129-5p; miR-1229-3p; and miR-1224-3p; in the regulation of mitochondrial function exhibited strong associations with type 2 diabetes. These results might aid in the creation of novel targets for therapy and type 2 diabetes biomarkers.
Collapse
Affiliation(s)
- Kritika Sharma
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, New Delhi 110007, India; Department of Biotechnology, Delhi Technological University, Delhi 110042, India
| | - Neeru Saini
- CSIR-Institute of Genomics and Integrative Biology, Mall Road, New Delhi 110007, India; Academy of Scientific & Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Yasha Hasija
- Department of Biotechnology, Delhi Technological University, Delhi 110042, India.
| |
Collapse
|
6
|
Rabby MG, Rahman MH, Islam MN, Kamal MM, Biswas M, Bonny M, Hasan MM. In silico identification and functional prediction of differentially expressed genes in South Asian populations associated with type 2 diabetes. PLoS One 2023; 18:e0294399. [PMID: 38096208 PMCID: PMC10721103 DOI: 10.1371/journal.pone.0294399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 11/01/2023] [Indexed: 12/17/2023] Open
Abstract
Type 2 diabetes (T2D) is one of the major metabolic disorders in humans caused by hyperglycemia and insulin resistance syndrome. Although significant genetic effects on T2D pathogenesis are experimentally proved, the molecular mechanism of T2D in South Asian Populations (SAPs) is still limited. Hence, the current research analyzed two Gene Expression Omnibus (GEO) and 17 Genome-Wide Association Studies (GWAS) datasets associated with T2D in SAP to identify DEGs (differentially expressed genes). The identified DEGs were further analyzed to explore the molecular mechanism of T2D pathogenesis following a series of bioinformatics approaches. Following PPI (Protein-Protein Interaction), 867 potential DEGs and nine hub genes were identified that might play significant roles in T2D pathogenesis. Interestingly, CTNNB1 and RUNX2 hub genes were found to be unique for T2D pathogenesis in SAPs. Then, the GO (Gene Ontology) showed the potential biological, molecular, and cellular functions of the DEGs. The target genes also interacted with different pathways of T2D pathogenesis. In fact, 118 genes (including HNF1A and TCF7L2 hub genes) were directly associated with T2D pathogenesis. Indeed, eight key miRNAs among 2582 significantly interacted with the target genes. Even 64 genes were downregulated by 367 FDA-approved drugs. Interestingly, 11 genes showed a wide range (9-43) of drug specificity. Hence, the identified DEGs may guide to elucidate the molecular mechanism of T2D pathogenesis in SAPs. Therefore, integrating the research findings of the potential roles of DEGs and candidate drug-mediated downregulation of marker genes, future drugs or treatments could be developed to treat T2D in SAPs.
Collapse
Affiliation(s)
- Md. Golam Rabby
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Khulna, Bangladesh
| | - Md. Hafizur Rahman
- Department of Agro Product Processing Technology, Jashore University of Science and Technology, Khulna, Bangladesh
- Faculty of Food Sciences and Safety, Department of Quality Control and Safety Management, Khulna Agricultural University, Khulna, Bangladesh
| | - Md. Numan Islam
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Khulna, Bangladesh
| | - Md. Mostafa Kamal
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Khulna, Bangladesh
| | - Mrityunjoy Biswas
- Department of Agro Product Processing Technology, Jashore University of Science and Technology, Khulna, Bangladesh
| | - Mantasa Bonny
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Khulna, Bangladesh
| | - Md. Mahmudul Hasan
- Department of Nutrition and Food Technology, Jashore University of Science and Technology, Khulna, Bangladesh
| |
Collapse
|
7
|
Slick RA, Tinklenberg JA, Sutton J, Zhang L, Meng H, Beatka MJ, Vanden Avond M, Prom MJ, Ott E, Montanaro F, Heisner J, Toro R, Granzier H, Geurts AM, Stowe DF, Hill RB, Lawlor MW. Aberrations in Energetic Metabolism and Stress-Related Pathways Contribute to Pathophysiology in the Neb Conditional Knockout Mouse Model of Nemaline Myopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2023; 193:1528-1547. [PMID: 37422147 PMCID: PMC10548278 DOI: 10.1016/j.ajpath.2023.06.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 05/08/2023] [Accepted: 06/08/2023] [Indexed: 07/10/2023]
Abstract
Nemaline myopathy (NM) is a genetically and clinically heterogeneous disease that is diagnosed on the basis of the presence of nemaline rods on skeletal muscle biopsy. Although NM has typically been classified by causative genes, disease severity or prognosis cannot be predicted. The common pathologic end point of nemaline rods (despite diverse genetic causes) and an unexplained range of muscle weakness suggest that shared secondary processes contribute to the pathogenesis of NM. We speculated that these processes could be identified through a proteome-wide interrogation using a mouse model of severe NM in combination with pathway validation and structural/functional analyses. A proteomic analysis was performed using skeletal muscle tissue from the Neb conditional knockout mouse model compared with its wild-type counterpart to identify pathophysiologically relevant biological processes that might impact disease severity or provide new treatment targets. A differential expression analysis and Ingenuity Pathway Core Analysis predicted perturbations in several cellular processes, including mitochondrial dysfunction and changes in energetic metabolism and stress-related pathways. Subsequent structural and functional studies demonstrated abnormal mitochondrial distribution, decreased mitochondrial respiratory function, an increase in mitochondrial transmembrane potential, and extremely low ATP content in Neb conditional knockout muscles relative to wild type. Overall, the findings of these studies support a role for severe mitochondrial dysfunction as a novel contributor to muscle weakness in NM.
Collapse
Affiliation(s)
- Rebecca A Slick
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jennifer A Tinklenberg
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Clinical and Translational Science Institute, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jessica Sutton
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Liwen Zhang
- Mass Spectrometry and Proteomics Facility, Campus Chemical Instrument Center, The Ohio State University, Columbus, Ohio
| | - Hui Meng
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Margaret J Beatka
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mark Vanden Avond
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Mariah J Prom
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Emily Ott
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Federica Montanaro
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Research and Teaching Department, UCL Great Ormond Street Institute of Child Health, London, United Kingdom the NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom; NIHR Great Ormond Street Hospital Biomedical Research Centre, London, United Kingdom
| | - James Heisner
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Rafael Toro
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Henk Granzier
- College of Medicine, University of Arizona, Tucson, Arizona
| | - Aron M Geurts
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Mellowes Center for Genomic Sciences and Precision Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - David F Stowe
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Anesthesiology, Medical College of Wisconsin, Milwaukee, Wisconsin; Joint Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, Wisconsin
| | - R Blake Hill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael W Lawlor
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine and Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin; Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin.
| |
Collapse
|
8
|
Whytock KL, Pino MF, Sun Y, Yu G, De Carvalho FG, Yeo RX, Vega RB, Parmar G, Divoux A, Kapoor N, Yi F, Cornnell H, Patten DA, Harper ME, Gardell SJ, Smith SR, Walsh MJ, Sparks LM. Comprehensive interrogation of human skeletal muscle reveals a dissociation between insulin resistance and mitochondrial capacity. Am J Physiol Endocrinol Metab 2023; 325:E291-E302. [PMID: 37584609 DOI: 10.1152/ajpendo.00143.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023]
Abstract
Insulin resistance and blunted mitochondrial capacity in skeletal muscle are often synonymous, however, this association remains controversial. The aim of this study was to perform an in-depth multifactorial comparison of skeletal muscle mitochondrial capacity between individuals who were lean and active (Active, n = 9), individuals with obesity (Obese, n = 9), and individuals with obesity, insulin resistance, and type 2 diabetes (T2D, n = 22). Mitochondrial capacity was assessed by ex vivo mitochondrial respiration with fatty-acid and glycolytic-supported protocols adjusted for mitochondrial content (mtDNA and citrate synthase activity). Supercomplex assembly was measured by Blue Native (BN)-PAGE and immunoblot. Tricarboxylic (TCA) cycle intermediates were assessed with targeted metabolomics. Exploratory transcriptomics and DNA methylation analyses were performed to uncover molecular differences affecting mitochondrial function among the three groups. We reveal no discernable differences in skeletal muscle mitochondrial content, mitochondrial capacity, supercomplex assembly, TCA cycle intermediates, and mitochondrial molecular profiles between obese individuals with and without T2D that had comparable levels of confounding factors (body mass index, age, and aerobic capacity). We highlight that lean, active individuals have greater mitochondrial content, mitochondrial capacity, supercomplex assembly, and TCA cycle intermediates. These phenotypical changes are reflected at the level of DNA methylation and gene transcription. The collective observation of comparable muscle mitochondrial capacity in individuals with obesity and T2D (vs. individuals without T2D) underscores a dissociation from skeletal muscle insulin resistance. Clinical trial number: NCT01911104.NEW & NOTEWORTHY Whether impaired mitochondrial capacity contributes to skeletal muscle insulin resistance is debated. Our multifactorial analysis shows no differences in skeletal muscle mitochondrial content, mitochondrial capacity, and mitochondrial molecular profiles between obese individuals with and without T2D that had comparable levels of confounding factors (BMI, age, aerobic capacity). We highlight that lean, active individuals have enhanced skeletal muscle mitochondrial capacity that is also reflected at the level of DNA methylation and gene transcription.
Collapse
Affiliation(s)
- Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Maria F Pino
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Yifei Sun
- Icahn School of Medicine at Mount Sinai, New York City, New York, United States
| | - GongXin Yu
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | | | - Reichelle X Yeo
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Rick B Vega
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Gaganvir Parmar
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Nidhi Kapoor
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Fancaho Yi
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Heather Cornnell
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - David A Patten
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Stephen J Gardell
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Martin J Walsh
- Icahn School of Medicine at Mount Sinai, New York City, New York, United States
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| |
Collapse
|
9
|
Hatton AA, Hillary RF, Bernabeu E, McCartney DL, Marioni RE, McRae AF. Blood-based genome-wide DNA methylation correlations across body-fat- and adiposity-related biochemical traits. Am J Hum Genet 2023; 110:1564-1573. [PMID: 37652023 PMCID: PMC10502853 DOI: 10.1016/j.ajhg.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 08/04/2023] [Accepted: 08/03/2023] [Indexed: 09/02/2023] Open
Abstract
The recent increase in obesity levels across many countries is likely to be driven by nongenetic factors. The epigenetic modification DNA methylation (DNAm) may help to explore this, as it is sensitive to both genetic and environmental exposures. While the relationship between DNAm and body-fat traits has been extensively studied, there is limited literature on the shared associations of DNAm variation across such traits. Akin to genetic correlation estimates, here, we introduce an approach to evaluate the similarities in DNAm associations between traits: DNAm correlations. As DNAm can be both a cause and consequence of complex traits, DNAm correlations have the potential to provide insights into trait relationships above that currently obtained from genetic and phenotypic correlations. Utilizing 7,519 unrelated individuals from Generation Scotland with DNAm from the EPIC array, we calculated DNAm correlations between body-fat- and adiposity-related traits by using the bivariate OREML framework in the OSCA software. For each trait, we also estimated the shared contribution of DNAm between sexes. We identified strong, positive DNAm correlations between each of the body-fat traits (BMI, body-fat percentage, and waist-to-hip ratio, ranging from 0.96 to 1.00), finding larger associations than those identified by genetic and phenotypic correlations. We identified a significant deviation from 1 in the DNAm correlations for BMI between males and females, with sex-specific DNAm changes associated with BMI identified at eight DNAm probes. Employing genome-wide DNAm correlations to evaluate the similarities in the associations of DNAm with complex traits has provided insight into obesity-related traits beyond that provided by genetic correlations.
Collapse
Affiliation(s)
| | - Robert F Hillary
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Elena Bernabeu
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Daniel L McCartney
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Riccardo E Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK
| | - Allan F McRae
- Institute for Molecular Bioscience, Brisbane, Australia.
| |
Collapse
|
10
|
Guo Y, Miao X, Sun X, Li L, Zhou A, Zhu X, Xu Y, Wang Q, Li Z, Fan Z. Zinc finger transcription factor Egf1 promotes non-alcoholic fatty liver disease. JHEP Rep 2023; 5:100724. [PMID: 37234276 PMCID: PMC10206499 DOI: 10.1016/j.jhepr.2023.100724] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/19/2023] [Accepted: 02/22/2023] [Indexed: 05/27/2023] Open
Abstract
Background & Aims Non-alcoholic fatty liver disease (NAFLD) contributes to the global epidemic of metabolic syndrome and is considered a prelude to end-stage liver diseases such as cirrhosis and hepatocellular carcinoma. During NAFLD pathogenesis, hepatic parenchymal cells (hepatocytes) undergo both morphological and functional changes owing to a rewired transcriptome. The underlying mechanism is not entirely clear. In the present study, we investigated the involvement of early growth response 1 (Egr1) in NAFLD. Methods Quantitative PCR, Western blotting, and histochemical staining were used to assess gene expression levels. Chromatin immunoprecipitation was used to evaluate protein binding to DNA. NAFLD was evaluated in leptin receptor-deficient (db/db) mice. Results We report here that Egr1 was upregulated by pro-NAFLD stimuli in vitro and in vivo. Further analysis revealed that serum response factor (SRF) was recruited to the Egr1 promoter and mediated Egr1 transactivation. Importantly, Egr1 depletion markedly mitigated NAFLD in db/db mice. RNA sequencing revealed that Egr1 knockdown in hepatocytes, on the one hand, boosted fatty acid oxidation (FAO) and, on the other hand, suppressed the synthesis of chemoattractants. Mechanistically, Egr1 interacted with peroxisome proliferator-activated receptor α (PPARα) to repress PPARα-dependent transcription of FAO genes by recruiting its co-repressor NGFI-A binding protein 1 (Nab1), which potentially led to promoter deacetylation of FAO genes. Conclusions Our data identify Egr1 as a novel modulator of NAFLD and a potential target for NAFLD intervention. Impact and Implications Non-alcoholic fatty liver disease (NAFLD) precedes cirrhosis and hepatocellular carcinoma. In this paper, we describe a novel mechanism whereby early growth response 1 (Egr1), a transcription factor, contributes to NAFLD pathogenesis by regulating fatty acid oxidation. Our data provide novel insights and translational potential for NAFLD intervention.
Collapse
Affiliation(s)
- Yan Guo
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Xiulian Miao
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Xinyue Sun
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Luyang Li
- Department of Oral Medicine, Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, China
| | - Anqi Zhou
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
| | - Xi Zhu
- Department of Infectious Diseases, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Yong Xu
- Institute of Biomedical Research and College of Life Sciences, Liaocheng University, Liaocheng, China
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Qinghua Wang
- Department of Gastroenterology, Kunshan First People's Hospital Affiliated to Jiangsu University, Kunshan, China
| | - Zilong Li
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing, China
| | - Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
11
|
Liu L, Sun K, Luo Y, Wang B, Yang Y, Chen L, Zheng S, Wu T, Xiao P. Myocardin-related transcription factor A, regulated by serum response factor, contributes to diabetic cardiomyopathy in mice. Life Sci 2023; 317:121470. [PMID: 36758668 DOI: 10.1016/j.lfs.2023.121470] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/27/2023] [Accepted: 01/29/2023] [Indexed: 02/10/2023]
Abstract
AIMS Diabetic cardiomyopathy is a significant contributor to the global pandemic of heart failure. In the present study we investigated the involvement of myocardin-related transcription factor A (MRTF-A), a transcriptional regulator, in this process. MATERIALS AND METHODS Diabetic cardiomyopathy was induced in mice by feeding with a high-fat diet (HFD) or streptozotocin (STZ) injection. KEY FINDINGS We report that MRTF-A was up-regulated in the hearts of mice with diabetic cardiomyopathy. MRTF-A expression was also up-regulated by treatment with palmitate in cultured cardiomyocytes in vitro. Mechanistically, serum response factor (SRF) bound to the MRTF-A gene promoter and activated MRTF-A transcription in response to pro-diabetic stimuli. Knockdown of SRF abrogated MRTF-A induction in cardiomyocytes treated with palmitate. When cardiomyocytes conditional MRTF-A knockout mice (MRTF-A CKO) and wild type (WT) mice were placed on an HFD to induce diabetic cardiomyopathy, it was found that the CKO mice and the WT mice displayed comparable metabolic parameters including body weight, blood insulin concentration, blood cholesterol concentration, and glucose tolerance. However, both systolic and diastolic cardiac function were exacerbated by MRTF-A deletion in the heart. SIGNIFICANCE These data suggest that MRTF-A up-regulation might serve as an important compensatory mechanism to safeguard the deterioration of cardiac function during diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Li Liu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China; Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China; Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Ke Sun
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Yajun Luo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Bingshu Wang
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Medical Research Center of The First Affiliated Hospital, Hainan Women and Children Medical Center, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou 571199, China; Department of Pathology, The Second Affiliated Hospital of Hainan Medical University, Haikou 570216, China
| | - Yuyu Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Long Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing 210023, China
| | - Shaojiang Zheng
- Key Laboratory of Tropical Cardiovascular Diseases Research of Hainan Province, Medical Research Center of The First Affiliated Hospital, Hainan Women and Children Medical Center, Key Laboratory of Emergency and Trauma of Ministry of Education, Hainan Medical University, Haikou 571199, China.
| | - Teng Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease and Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China.
| | - Pingxi Xiao
- Department of Cardiology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
12
|
Jones RG, Dimet-Wiley A, Haghani A, da Silva FM, Brightwell CR, Lim S, Khadgi S, Wen Y, Dungan CM, Brooke RT, Greene NP, Peterson CA, McCarthy JJ, Horvath S, Watowich SJ, Fry CS, Murach KA. A molecular signature defining exercise adaptation with ageing and in vivo partial reprogramming in skeletal muscle. J Physiol 2023; 601:763-782. [PMID: 36533424 PMCID: PMC9987218 DOI: 10.1113/jp283836] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Exercise promotes functional improvements in aged tissues, but the extent to which it simulates partial molecular reprogramming is unknown. Using transcriptome profiling from (1) a skeletal muscle-specific in vivo Oct3/4, Klf4, Sox2 and Myc (OKSM) reprogramming-factor expression murine model; (2) an in vivo inducible muscle-specific Myc induction murine model; (3) a translatable high-volume hypertrophic exercise training approach in aged mice; and (4) human exercise muscle biopsies, we collectively defined exercise-induced genes that are common to partial reprogramming. Late-life exercise training lowered murine DNA methylation age according to several contemporary muscle-specific clocks. A comparison of the murine soleus transcriptome after late-life exercise training to the soleus transcriptome after OKSM induction revealed an overlapping signature that included higher JunB and Sun1. Also, within this signature, downregulation of specific mitochondrial and muscle-enriched genes was conserved in skeletal muscle of long-term exercise-trained humans; among these was muscle-specific Abra/Stars. Myc is the OKSM factor most induced by exercise in muscle and was elevated following exercise training in aged mice. A pulse of MYC rewired the global soleus muscle methylome, and the transcriptome after a MYC pulse partially recapitulated OKSM induction. A common signature also emerged in the murine MYC-controlled and exercise adaptation transcriptomes, including lower muscle-specific Melusin and reactive oxygen species-associated Romo1. With Myc, OKSM and exercise training in mice, as well habitual exercise in humans, the complex I accessory subunit Ndufb11 was lower; low Ndufb11 is linked to longevity in rodents. Collectively, exercise shares similarities with genetic in vivo partial reprogramming. KEY POINTS: Advances in the last decade related to cellular epigenetic reprogramming (e.g. DNA methylome remodelling) toward a pluripotent state via the Yamanaka transcription factors Oct3/4, Klf4, Sox2 and Myc (OKSM) provide a window into potential mechanisms for combatting the deleterious effects of cellular ageing. Using global gene expression analysis, we compared the effects of in vivo OKSM-mediated partial reprogramming in skeletal muscle fibres of mice to the effects of late-life murine exercise training in muscle. Myc is the Yamanaka factor most induced by exercise in skeletal muscle, and so we compared the MYC-controlled transcriptome in muscle to Yamanaka factor-mediated and exercise adaptation mRNA landscapes in mice and humans. A single pulse of MYC is sufficient to remodel the muscle methylome. We identify partial reprogramming-associated genes that are innately altered by exercise training and conserved in humans, and propose that MYC contributes to some of these responses.
Collapse
Affiliation(s)
- Ronald G. Jones
- University of Arkansas, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
| | | | - Amin Haghani
- University of California Los Angeles, Department of Human Genetics, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Francielly Morena da Silva
- University of Arkansas, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
- University of Arkansas, Cachexia Research Laboratory, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
| | - Camille R. Brightwell
- University of Kentucky Center for Muscle Biology, Lexington, KY, USA
- University of Kentucky, Department of Athletic Training and Clinical Nutrition, Lexington, KY, USA
| | - Seongkyun Lim
- University of Arkansas, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
- University of Arkansas, Cachexia Research Laboratory, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
| | - Sabin Khadgi
- University of Arkansas, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
| | - Yuan Wen
- University of Kentucky Center for Muscle Biology, Lexington, KY, USA
- University of Kentucky, Department of Physical Therapy, Lexington, KY, USA
| | - Cory M. Dungan
- University of Kentucky Center for Muscle Biology, Lexington, KY, USA
- University of Kentucky, Department of Physical Therapy, Lexington, KY, USA
| | | | - Nicholas P. Greene
- University of Arkansas, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
- University of Arkansas, Cachexia Research Laboratory, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
- University of Arkansas, Cell and Molecular Biology Graduate Program, Fayetteville, AR, USA
| | - Charlotte A. Peterson
- University of Kentucky Center for Muscle Biology, Lexington, KY, USA
- University of Kentucky, Department of Physical Therapy, Lexington, KY, USA
- University of Kentucky, Department of Physiology, Lexington, KY, USA
| | - John J. McCarthy
- Altos Labs, San Diego, CA, USA
- University of Kentucky, Department of Physiology, Lexington, KY, USA
| | - Steve Horvath
- University of California Los Angeles, Department of Human Genetics, Los Angeles, CA, USA
- Altos Labs, San Diego, CA, USA
| | - Stanley J. Watowich
- Ridgeline Therapeutics, Houston, TX, USA
- University of Texas Medical Branch, Department of Biochemistry and Molecular Biology, Galveston, TX, USA
| | - Christopher S. Fry
- University of Kentucky Center for Muscle Biology, Lexington, KY, USA
- University of Kentucky, Department of Athletic Training and Clinical Nutrition, Lexington, KY, USA
| | - Kevin A. Murach
- University of Arkansas, Exercise Science Research Center, Department of Health, Human Performance, and Recreation, Fayetteville, AR, USA
- University of Arkansas, Cell and Molecular Biology Graduate Program, Fayetteville, AR, USA
| |
Collapse
|
13
|
Rho/SRF Inhibitor Modulates Mitochondrial Functions. Int J Mol Sci 2022; 23:ijms231911536. [PMID: 36232837 PMCID: PMC9570101 DOI: 10.3390/ijms231911536] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 12/04/2022] Open
Abstract
CCG-1423 is a Rho A pathway inhibitor that has been reported to inhibit Rho/SRF-mediated transcriptional regulation. Serum response factor and its cofactors, which include ternary complex factors and myocardin-related transcription factors, regulate various cellular functions. In this study, we observed that CCG-1423 modulates the mitochondrial functions. The effect of this small molecule drug was determined by measuring mitochondrial function using an XFe96 Analyzer and an Oxygraph 2k (O2k) high-resolution respirometer. CCG-1423 treatment significantly reduced oxidative phosphorylation in a dose-dependent manner. However, CCG-1423 increased the glycolytic rate. We also observed that histone 4 at lysine-16 underwent hyperacetylation with the treatment of this drug. Immunolabeling with F-actin and MitoTracker revealed the alteration in the actin cytoskeleton and mitochondria. Taken together, our findings highlight a critical role of CCG-1423 in inhibiting the transcription of SRF/p49 and PGC-1α, β, resulting in the downregulation of mitochondrial genes, leading to the repression of mitochondrial oxidative phosphorylation and overall ATP reduction. This study provides a better understanding of the effects of CCG-1423 on mitochondria, which may be useful for the assessment of the potential clinical application of CCG-1423 and its derivatives.
Collapse
|
14
|
Galindo CL, Nguyen VT, Hill B, Easterday E, Cleator JH, Sawyer DB. Neuregulin (NRG-1β) Is Pro-Myogenic and Anti-Cachectic in Respiratory Muscles of Post-Myocardial Infarcted Swine. BIOLOGY 2022; 11:682. [PMID: 35625411 PMCID: PMC9137990 DOI: 10.3390/biology11050682] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/30/2022]
Abstract
Neuregulin-1β (NRG-1β) is a growth and differentiation factor with pleiotropic systemic effects. Because NRG-1β has therapeutic potential for heart failure and has known growth effects in skeletal muscle, we hypothesized that it might affect heart failure-associated cachexia, a severe co-morbidity characterized by a loss of muscle mass. We therefore assessed NRG-1β's effect on intercostal skeletal muscle gene expression in a swine model of heart failure using recombinant glial growth factor 2 (USAN-cimaglermin alfa), a version of NRG-1β that has been tested in humans with systolic heart failure. Animals received one of two intravenous doses (0.67 or 2 mg/kg) of NRG-1β bi-weekly for 4 weeks, beginning one week after infarct. Based on paired-end RNA sequencing, NRG-1β treatment altered the intercostal muscle gene expression of 581 transcripts, including genes required for myofiber growth, maintenance and survival, such as MYH3, MYHC, MYL6B, KY and HES1. Importantly, NRG-1β altered the directionality of at least 85 genes associated with cachexia, including myostatin, which negatively regulates myoblast differentiation by down-regulating MyoD expression. Consistent with this, MyoD was increased in NRG-1β-treated animals. In vitro experiments with myoblast cell lines confirmed that NRG-1β induces ERBB-dependent differentiation. These findings suggest a NRG-1β-mediated anti-atrophic, anti-cachexia effect that may provide additional benefits to this potential therapy in heart failure.
Collapse
Affiliation(s)
- Cristi L. Galindo
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - Van Thuan Nguyen
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - Braxton Hill
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - Ethan Easterday
- Department of Biology, Ogden College of Science & Engineering, Western Kentucky University, Bowling Green, KY 42101, USA; (V.T.N.); (B.H.); (E.E.)
| | - John H. Cleator
- Centennial Heart at Skyline, 3443 Dickerson Pike, Suite 430, Nashville, TN 37207, USA;
| | - Douglas B. Sawyer
- Department of Cardiac Services, Maine Medical Center, Scarborough, ME 04074, USA
| |
Collapse
|
15
|
Liu S, Cai X, Wang T, Xu J, Cheng W, Wang X, Wei G, Yan S. Downregulation of ALDH6A1 is a New Marker of Muscle Insulin Resistance in Type 2 Diabetes Mellitus. Int J Gen Med 2022; 15:2137-2147. [PMID: 35241929 PMCID: PMC8887615 DOI: 10.2147/ijgm.s343727] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 01/21/2022] [Indexed: 12/23/2022] Open
Abstract
Purpose Skeletal muscle insulin resistance (IR) is an important etiology of type 2 diabetes mellitus (T2DM); however, its molecular mechanism is yet to be fully defined. This study attempted to identify the gene expression patterns and molecular disorders in T2DM patients’ skeletal muscle samples. Methods First, the difference in genetic expression among GSE25462 data was analyzed. Next, PPI network analysis of differential genes was carried out, after which the maladjustment module was identified. Then, an enrichment analysis and gene set enrichment analysis (GSEA) were carried out. Finally, the transcription factors that regulate the modular genes by raid were predicted. Results Most differentially expressed genes were found to be able to form an interaction network and cluster into 9 modules. These modular genes were shown to possess a significant correlation with immune inflammation and metabolic response. Importantly, the top 15 genes of area under receiver operating characteristic curve (AUC) were identified, and the expression of 10 genes by GSE12643, GSE18732 and GSE29221 was confirmed. The expression and AUC value of ALDH6A1 were then verified according to three sets of data, where ALDH6A1 was found to be negatively correlated with follicular helper T cells. However, among the predicted transcription regulators, HDAC was shown to have a better regulatory effect. Conclusion The findings highlight that the dysregulation of ALDH6A1 expression in IR of T2DM may serve as a potential therapeutic target. ALDH6A1 is involved in the immune inflammation and metabolic pathways.
Collapse
Affiliation(s)
- Song Liu
- Endocrinology Department, Traditional Chinese Medicine Academy of Heilongjiang, Harbin, Heilongjiang Province, People’s Republic of China
| | - Xiaojun Cai
- Endocrinology Department, Traditional Chinese Medicine Academy of Heilongjiang, Harbin, Heilongjiang Province, People’s Republic of China
| | - Tao Wang
- Endocrinology Department, Traditional Chinese Medicine Academy of Heilongjiang, Harbin, Heilongjiang Province, People’s Republic of China
| | - Jiwen Xu
- Anatomy, Histology and Embryology Teaching and Research Section, School of Basic Medicine, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang Province, People’s Republic of China
| | - Weilun Cheng
- Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
| | - Xuling Wang
- Endocrinology Department, Traditional Chinese Medicine Academy of Heilongjiang, Harbin, Heilongjiang Province, People’s Republic of China
| | - Gangjie Wei
- Medical Department, Traditional Chinese Medicine Academy of Heilongjiang, Harbin, Heilongjiang Province, People’s Republic of China
| | - Shuang Yan
- Department of Endocrinology and Metabolism, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, People’s Republic of China
- Correspondence: Shuang Yan, Email
| |
Collapse
|
16
|
Ravi S, Gunawan R. ΔFBA-Predicting metabolic flux alterations using genome-scale metabolic models and differential transcriptomic data. PLoS Comput Biol 2021; 17:e1009589. [PMID: 34758020 PMCID: PMC8608322 DOI: 10.1371/journal.pcbi.1009589] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 11/22/2021] [Accepted: 10/25/2021] [Indexed: 12/04/2022] Open
Abstract
Genome-scale metabolic models (GEMs) provide a powerful framework for simulating the entire set of biochemical reactions in a cell using a constraint-based modeling strategy called flux balance analysis (FBA). FBA relies on an assumed metabolic objective for generating metabolic fluxes using GEMs. But, the most appropriate metabolic objective is not always obvious for a given condition and is likely context-specific, which often complicate the estimation of metabolic flux alterations between conditions. Here, we propose a new method, called ΔFBA (deltaFBA), that integrates differential gene expression data to evaluate directly metabolic flux differences between two conditions. Notably, ΔFBA does not require specifying the cellular objective. Rather, ΔFBA seeks to maximize the consistency and minimize inconsistency between the predicted flux differences and differential gene expression. We showcased the performance of ΔFBA through several case studies involving the prediction of metabolic alterations caused by genetic and environmental perturbations in Escherichia coli and caused by Type-2 diabetes in human muscle. Importantly, in comparison to existing methods, ΔFBA gives a more accurate prediction of flux differences. Metabolic alterations are often used as hallmarks of observable phenotypes. In this regard, reconstructed genome-scale metabolic models (GEMs) provide a rich and computable representation of the entire set of biochemical reactions in a cell. However, the performance of analytical tools for predicting metabolic reaction rates or fluxes using GEMs is sensitive to the assumed metabolic objective that is often unknown and likely context-specific. Here, we propose a novel method called ΔFBA that combines differential gene expression data and GEMs to evaluate differences in the metabolic fluxes between two conditions (perturbation vs. control) without the need for specifying a metabolic objective. In our demonstration, ΔFBA outperformed other existing methods in predicting metabolic flux alterations.
Collapse
Affiliation(s)
- Sudharshan Ravi
- Department of Chemical and Biological Engineering, University at Buffalo-SUNY, Buffalo, New York, United States of America
- Institute for Chemical and Bioengineering, ETH Zurich, Zurich, Switzerland
| | - Rudiyanto Gunawan
- Department of Chemical and Biological Engineering, University at Buffalo-SUNY, Buffalo, New York, United States of America
- * E-mail:
| |
Collapse
|
17
|
Yurista SR, Silljé HHW, Nijholt KT, Dokter MM, van Veldhuisen DJ, de Boer RA, Westenbrink BD. Factor Xa Inhibition with Apixaban Does Not Influence Cardiac Remodelling in Rats with Heart Failure After Myocardial Infarction. Cardiovasc Drugs Ther 2021; 35:953-963. [PMID: 32458320 PMCID: PMC8452585 DOI: 10.1007/s10557-020-06999-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heart failure (HF) is considered to be a prothrombotic condition and it has been suggested that coagulation factors contribute to maladaptive cardiac remodelling via activation of the protease-activated receptor 1 (PAR1). We tested the hypothesis that anticoagulation with the factor Xa (FXa) inhibitor apixaban would ameliorate cardiac remodelling in rats with HF after myocardial infarction (MI). METHODS AND RESULTS Male Sprague-Dawley rats were either subjected to permanent ligation of the left ascending coronary artery (MI) or sham surgery. The MI and sham animals were randomly allocated to treatment with placebo or apixaban in the chow (150 mg/kg/day), starting 2 weeks after surgery. Cardiac function was assessed using echocardiography and histological and molecular markers of cardiac hypertrophy were assessed in the left ventricle (LV). Apixaban resulted in a fivefold increase in anti-FXa activity compared with vehicle, but no overt bleeding was observed and haematocrit levels remained similar in apixaban- and vehicle-treated groups. After 10 weeks of treatment, LV ejection fraction was 42 ± 3% in the MI group treated with apixaban and 37 ± 2 in the vehicle-treated MI group (p > 0.05). Both vehicle- and apixaban-treated MI groups also displayed similar degrees of LV dilatation, LV hypertrophy and interstitial fibrosis. Histological and molecular markers for pathological remodelling were also comparable between groups, as was the activity of signalling pathways downstream of the PAR1 receptor. CONCLUSION FXa inhibition with apixaban does not influence pathological cardiac remodelling after MI. These data do not support the use of FXa inhibitor in HF patients with the aim to amend the severity of HF. Graphical Abstract.
Collapse
Affiliation(s)
- Salva R Yurista
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen, 9700 RB, The Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen, 9700 RB, The Netherlands
| | - Kirsten T Nijholt
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen, 9700 RB, The Netherlands
| | - Martin M Dokter
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen, 9700 RB, The Netherlands
| | - Dirk J van Veldhuisen
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen, 9700 RB, The Netherlands
| | - Rudolf A de Boer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen, 9700 RB, The Netherlands
| | - B Daan Westenbrink
- Department of Cardiology, University Medical Center Groningen, University of Groningen, PO Box 30.001, Groningen, 9700 RB, The Netherlands.
| |
Collapse
|
18
|
Shinohara M, Kikuchi M, Onishi-Takeya M, Tashiro Y, Suzuki K, Noda Y, Takeda S, Mukouzono M, Nagano S, Fukumori A, Morishita R, Nakaya A, Sato N. Upregulated expression of a subset of genes in APP; ob/ ob mice: Evidence of an interaction between diabetes-linked obesity and Alzheimer's disease. FASEB Bioadv 2021; 3:323-333. [PMID: 33977233 PMCID: PMC8103720 DOI: 10.1096/fba.2020-00151] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/17/2021] [Accepted: 01/29/2021] [Indexed: 12/26/2022] Open
Abstract
Clinical studies have indicated that obesity and diabetes are associated with Alzheimer's disease (AD) and neurodegeneration. However, the mechanism by which obesity/diabetes and AD interact with each other and contribute to dementia remains elusive. To obtain insights into their interaction at molecular levels, we performed gene expression analysis of APP;ob/ob mice, which were generated by crossing transgenic AD model mice (APP23 mice) with ob/ob mice, which are obese and mildly diabetic. The Aβ level in these mice was reduced compared with that in pure APP mice. However, we identified a cluster of genes (cluster 10) upregulated in APP;ob/ob mice but not in either APP or ob/ob mice. Interestingly, genes upregulated in the human AD brain were enriched in cluster 10. Moreover, genes in cluster 10 formed a network and shared upregulated genes with a cell model of neurodegeneration and other models of neurological disorders such as ischemia and epilepsy. In silico analyses showed that serum response factor (SRF), recently identified in a single-cell analysis of human brains as a transcription factor that can control the conversion from healthy cells to AD cells, might be a common transcriptional regulator for a subset of cluster 10 genes. These data suggest that upregulation of genes uniquely associated with APP;ob/ob mice is an evidence of the interaction between obesity/diabetes and AD.
Collapse
Affiliation(s)
- Mitsuru Shinohara
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| | - Masataka Kikuchi
- Department of Genome Informatics Graduate School of Medicine Osaka University Osaka Japan
| | - Miyuki Onishi-Takeya
- Department of Geriatric Medicine Graduate School of Medicine Osaka University Osaka Japan
| | - Yoshitaka Tashiro
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Kaoru Suzuki
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Yasuhiro Noda
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan
| | - Shuko Takeda
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Masahiro Mukouzono
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Seiichi Nagano
- Department of Neurology Graduate School of Medicine Osaka University Osaka Japan
| | - Akio Fukumori
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| | - Ryuichi Morishita
- Department of Clinical Gene Therapy Graduate School of Medicine Osaka University Osaka Japan
| | - Akihiro Nakaya
- Department of Genome Informatics Graduate School of Medicine Osaka University Osaka Japan
| | - Naoyuki Sato
- Department of Aging Neurobiology Center for Development of Advanced Medicine for Dementia National Center for Geriatrics and Gerontology Obu Aichi Japan.,Department of Aging Neurobiology Graduate School of Medicine Osaka University Osaka Japan
| |
Collapse
|
19
|
Luo Y, Cui C, Han X, Wang Q, Zhang C. The role of miRNAs in polycystic ovary syndrome with insulin resistance. J Assist Reprod Genet 2021; 38:289-304. [PMID: 33405004 PMCID: PMC7884539 DOI: 10.1007/s10815-020-02019-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 11/22/2020] [Indexed: 02/07/2023] Open
Abstract
PURPOSE This review aims to summarize the key findings of several miRNAs and their roles in polycystic ovary syndrome with insulin resistance, characterize the disease pathogenesis, and establish a new theoretical basis for diagnosing, treating, and preventing polycystic ovary syndrome. METHODS Relevant scientific literature was covered from 1992 to 2020 by searching the PubMed database with search terms: insulin/insulin resistance, polycystic ovary syndrome, microRNAs, and metabolic diseases. References of relevant studies were cross-checked. RESULTS The related miRNAs (including differentially expressed miRNAs) and their roles in pathogenesis, and possible therapeutic targets and pathways, are discussed, highlighting controversies and offering thoughts for future directions. CONCLUSION We found abundant evidence on the role of differentially expressed miRNAs with its related phenotypes in PCOS. Considering the essential role of insulin resistance in the pathogenesis of PCOS, the alterations of associated miRNAs need more research attention. We speculate that race/ethnicity or PCOS phenotype and differences in methodological differences might lead to inconsistencies in research findings; thus, several miRNA profiles need to be investigated further to qualify for the potential therapeutic targets for PCOS-IR.
Collapse
Affiliation(s)
- Yingliu Luo
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Chenchen Cui
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Xiao Han
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Qian Wang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China
- Henan Joint International Research Laboratory of Reproductive Bioengineering, Zhengzhou, 450003, Henan Province, People's Republic of China
| | - Cuilian Zhang
- Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, 450003, Henan Province, People's Republic of China.
| |
Collapse
|
20
|
Maude H, Lau W, Maniatis N, Andrew T. New Insights Into Mitochondrial Dysfunction at Disease Susceptibility Loci in the Development of Type 2 Diabetes. Front Endocrinol (Lausanne) 2021; 12:694893. [PMID: 34456865 PMCID: PMC8385132 DOI: 10.3389/fendo.2021.694893] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 07/08/2021] [Indexed: 12/25/2022] Open
Abstract
This study investigated the potential genetic mechanisms which underlie adipose tissue mitochondrial dysfunction in Type 2 diabetes (T2D), by systematically identifying nuclear-encoded mitochondrial genes (NEMGs) among the genes regulated by T2D-associated genetic loci. The target genes of these 'disease loci' were identified by mapping genetic loci associated with both disease and gene expression levels (expression quantitative trait loci, eQTL) using high resolution genetic maps, with independent estimates co-locating to within a small genetic distance. These co-locating signals were defined as T2D-eQTL and the target genes as T2D cis-genes. In total, 763 cis-genes were associated with T2D-eQTL, of which 50 were NEMGs. Independent gene expression datasets for T2D and insulin resistant cases and controls confirmed that the cis-genes and cis-NEMGs were enriched for differential expression in cases, providing independent validation that genetic maps can identify informative functional genes. Two additional results were consistent with a potential role of T2D-eQTL in regulating the 50 identified cis-NEMGs in the context of T2D risk: (1) the 50 cis-NEMGs showed greater differential expression compared to other NEMGs and (2) other NEMGs showed a trend towards significantly decreased expression if their expression levels correlated more highly with the subset of 50 cis-NEMGs. These 50 cis-NEMGs, which are differentially expressed and associated with mapped T2D disease loci, encode proteins acting within key mitochondrial pathways, including some of current therapeutic interest such as the metabolism of branched-chain amino acids, GABA and biotin.
Collapse
Affiliation(s)
- Hannah Maude
- Department of Metabolism, Digestion & Reproduction, Imperial College, London, United Kingdom
| | - Winston Lau
- Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Nikolas Maniatis
- Department of Genetics, Evolution and Environment, University College London, London, United Kingdom
| | - Toby Andrew
- Department of Metabolism, Digestion & Reproduction, Imperial College, London, United Kingdom
- *Correspondence: Toby Andrew,
| |
Collapse
|
21
|
Watson RW, Azam H, Aura C, Russell N, McCormack J, Corey E, Morrissey C, Crown J, Gallagher WM, Prencipe M. Inhibition of Serum Response Factor Improves Response to Enzalutamide in Prostate Cancer. Cancers (Basel) 2020; 12:cancers12123540. [PMID: 33260953 PMCID: PMC7760758 DOI: 10.3390/cancers12123540] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 01/29/2023] Open
Abstract
Castrate-resistant prostate cancer (CRPC) is challenging to treat with the androgen receptor (AR), the main target and key focus of resistance. Understanding the mechanisms of AR interaction with co-regulators will identify new therapeutic targets to overcome AR resistance mechanisms. We previously identified the serum response factor (SRF) as a lead target in an in vitro model of CRPC and showed that SRF expression in tissues of CRPC patients was associated with shorter survival. Here, we tested SRF inhibition in vitro and in vivo to assess SRF as a potential target in CRPC. Inhibition of SRF with the small-molecule inhibitor CCG1423 resulted in enhanced response to enzalutamide in vitro and reduced tumour volume of LuCaP 35CR, a CRPC patient-derived xenograft model. Nuclear localisation of AR post-CCG1423 was significantly decreased and was associated with decreased α-tubulin acetylation in vitro and decreased prostate specific antigen (PSA) levels in vivo. SRF immunoreactivity was tested in metastatic tissues from CRPC patients to investigate its role in enzalutamide response. Kaplan-Meier curves showed that high SRF expression was associated with shorter response to enzalutamide. Our study supports the use of SRF inhibitors to improve response to enzalutamide.
Collapse
Affiliation(s)
- R. William Watson
- Conway Institute of Biomolecular and Biomedical Research, UCD School of Medicine, University College Dublin, Belfield, D4, Dublin, Ireland;
| | - Haleema Azam
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland; (H.A.); (C.A.); (N.R.); (W.M.G.)
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - Claudia Aura
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland; (H.A.); (C.A.); (N.R.); (W.M.G.)
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - Niamh Russell
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland; (H.A.); (C.A.); (N.R.); (W.M.G.)
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - Janet McCormack
- Research Pathology Core, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield D4, Dublin, Ireland;
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA 98195, USA; (E.C.); (C.M.)
| | - Colm Morrissey
- Department of Urology, University of Washington, Seattle, WA 98195, USA; (E.C.); (C.M.)
| | - John Crown
- Department of Medical Oncology, St Vincent’s University Hospital, Dublin, Ireland;
| | - William M Gallagher
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland; (H.A.); (C.A.); (N.R.); (W.M.G.)
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
| | - Maria Prencipe
- Cancer Biology and Therapeutics Laboratory, UCD Conway Institute, University College Dublin, Belfield, D4, Dublin, Ireland; (H.A.); (C.A.); (N.R.); (W.M.G.)
- UCD School of Biomolecular and Biomedical Science, University College Dublin, Belfield, D4, Dublin, Ireland
- Correspondence:
| |
Collapse
|
22
|
A hypothesis for insulin resistance in primary human adipocytes involving MRTF-A and suppression of PPARγ. Biochem Biophys Res Commun 2020; 533:64-69. [PMID: 32921413 DOI: 10.1016/j.bbrc.2020.08.105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 12/12/2022]
Abstract
Obesity is the main risk factor behind insulin resistance and type 2 diabetes. Still, the mechanism behind adipocyte dysfunction is not yet resolved. Recently, we reported that rapid actin remodeling correlates with adipose cell size changes after short-term overfeeding. Therefore, we hypothesized that the actin-driven myocardin-related transcription factor (MRTF-A) contributes to impaired mature adipocyte function. Primary human adipocytes were subjected to adenoviral overexpression of MRTF-A or MRTF-B, followed by Western blot analysis and tracer glucose uptake assay. Further, we assessed cell size distribution, insulin response, MRTF-A localization, actin organization and degree of polymerization in adipocytes isolated from Ob/Ob mice. Overexpression of MRTF-A, but not MRTF-B, markedly suppressed PPARγ expression. Further, MRTF-A expression resulted in decreased IRS-1 level, shifted phosphorylation of Akt (pS473/pT308), IRS-1 (pS302) and AS160 (pT642), and lowered insulin-stimulated glucose uptake. Hypertrophic adipocytes from Ob/Ob mice displayed an increased proportion of polymerized actin, and increased nuclear translocation of MRTF-A compared with control (Ob/+). Similar with human adipocytes overexpressing MRTF-A, adipocytes isolated from Ob/Ob mice had reduced expression of IRS-1 and PPARγ, as well as impaired insulin response. Together, these data demonstrate that MRTF-A negatively influences insulin sensitivity and the expression of key targets in fully mature human adipocytes. This suggests that MRTF-A is poised to exert a transcriptional response in hypertrophic adipocytes, contributing to adipocyte dysfunction and insulin resistance.
Collapse
|
23
|
Stern C, Schreier B, Nolze A, Rabe S, Mildenberger S, Gekle M. Knockout of vascular smooth muscle EGF receptor in a mouse model prevents obesity-induced vascular dysfunction and renal damage in vivo. Diabetologia 2020; 63:2218-2234. [PMID: 32548701 PMCID: PMC7476975 DOI: 10.1007/s00125-020-05187-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Obesity causes type 2 diabetes leading to vascular dysfunction and finally renal end-organ damage. Vascular smooth muscle (VSM) EGF receptor (EGFR) modulates vascular wall homeostasis in part via serum response factor (SRF), a major regulator of VSM differentiation and a sensor for glucose. We investigated the role of VSM-EGFR during obesity-induced renovascular dysfunction, as well as EGFR-hyperglycaemia crosstalk. METHODS The role of VSM-EGFR during high-fat diet (HFD)-induced type 2 diabetes was investigated in a mouse model with inducible, VSM-specific EGFR-knockout (KO). Various structural and functional variables as well as transcriptome changes, in vivo and ex vivo, were assessed. The impact of hyperglycaemia on EGFR-induced signalling and SRF transcriptional activity and the underlying mechanisms were investigated at the cellular level. RESULTS We show that VSM-EGFR mediates obesity/type 2 diabetes-induced vascular dysfunction, remodelling and transcriptome dysregulation preceding renal damage and identify an EGFR-glucose synergism in terms of SRF activation, matrix dysregulation and mitochondrial function. EGFR deletion protects the animals from HFD-induced endothelial dysfunction, creatininaemia and albuminuria. Furthermore, we show that HFD leads to marked changes of the aortic transcriptome in wild-type but not in KO animals, indicative of EGFR-dependent SRF activation, matrix dysregulation and mitochondrial dysfunction, the latter confirmed at the cellular level. Studies at the cellular level revealed that high glucose potentiated EGFR/EGF receptor 2 (ErbB2)-induced stimulation of SRF activity, enhancing the graded signalling responses to EGF, via the EGFR/ErbB2-ROCK-actin-MRTF pathway and promoted mitochondrial dysfunction. CONCLUSIONS/INTERPRETATION VSM-EGFR contributes to HFD-induced vascular and subsequent renal alterations. We propose that a potentiated EGFR/ErbB2-ROCK-MRTF-SRF signalling axis and mitochondrial dysfunction underlie the role of EGFR. This advanced working hypothesis will be investigated in mechanistic depth in future studies. VSM-EGFR may be a therapeutic target in cases of type 2 diabetes-induced renovascular disease. DATA AVAILABILITY The datasets generated during and/or analysed during the current study are available in: (1) share_it, the data repository of the academic libraries of Saxony-Anhalt ( https://doi.org/10.25673/32049.2 ); and (2) in the gene expression omnibus database with the study identity GSE144838 ( https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE144838 ). Graphical abstract.
Collapse
Affiliation(s)
- Christian Stern
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Barbara Schreier
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Alexander Nolze
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Sindy Rabe
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Sigrid Mildenberger
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany
| | - Michael Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Strasse 6, 06112, Halle, Germany.
| |
Collapse
|
24
|
Liu R, Xiong X, Nam D, Yechoor V, Ma K. SRF-MRTF signaling suppresses brown adipocyte development by modulating TGF-β/BMP pathway. Mol Cell Endocrinol 2020; 515:110920. [PMID: 32603734 PMCID: PMC7484394 DOI: 10.1016/j.mce.2020.110920] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/05/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022]
Abstract
The SRF/MRTF and upstream signaling cascade play key roles in actin cytoskeleton organization and myocyte development. To date, how this signaling axis may function in brown adipocyte lineage commitment and maturation has not been delineated. Here we report that MRTF-SRF signaling exerts inhibitory actions on brown adipogenesis, and suppressing this negative regulation promotes brown adipocyte lineage development. During brown adipogenic differentiation, protein expressions of SRF, MRTFA/B and its transcription targets were down-regulated, and MRTFA/B shuttled from nucleus to cytoplasm. Silencing of SRF or MRTF-A/MRTF-B enhanced two distinct stages of brown adipocyte development, mesenchymal stem cell determination to brown adipocytes and terminal differentiation of brown adipogenic progenitors. We further demonstrate that the MRTF-SRF axis exerts transcriptional regulations of the TGF-β and BMP signaling pathway, critical developmental cues for brown adipocyte development. TGF-β signaling activity was significantly attenuated, whereas that of the BMP pathway augmented by inhibition of SRF or MRTF-A/MRTF-B, leading to enhanced brown adipocyte differentiation. Our study demonstrates the MRTF-SRF transcriptional cascade as a negative regulator of brown adipogenesis, through its transcriptional control of the TGF-β/BMP signaling pathways.
Collapse
Affiliation(s)
- Ruya Liu
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA.
| | - Xuekai Xiong
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA
| | - Deokhwa Nam
- Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Vijay Yechoor
- Diabetes and Beta Cell Biology Center, Division of Endocrinology, Diabetes & Metabolism, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, 15213, USA
| | - Ke Ma
- Department of Diabetes Complications & Metabolism, Beckman Research Institute of City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
25
|
Parmer C, De Sousa-Coelho AL, Cheng HS, Daher G, Burkart A, Dreyfuss JM, Pan H, Prenner JC, Keilson JM, Pande R, Henkin S, Feinberg MW, Patti ME, Creager MA. Skeletal muscle expression of adipose-specific phospholipase in peripheral artery disease. Vasc Med 2020; 25:401-410. [PMID: 32853041 DOI: 10.1177/1358863x20947467] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Flow-limiting atherosclerotic lesions of arteries supplying the limbs are a cause of symptoms in patients with peripheral artery disease (PAD). Musculoskeletal metabolic factors also contribute to the pathophysiology of claudication, which is manifest as leg discomfort that impairs walking capacity. Accordingly, we conducted a case-control study to determine whether skeletal muscle metabolic gene expression is altered in PAD. Calf skeletal muscle gene expression of patients with PAD and healthy subjects was analyzed using microarrays. The top-ranking gene differentially expressed between PAD and controls (FDR < 0.001) was PLA2G16, which encodes adipose-specific phospholipase A2 (AdPLA) and is implicated in the maintenance of insulin sensitivity and regulation of lipid metabolism. Differential expression was confirmed by qRT-PCR; PLA2G16 was downregulated by 68% in patients with PAD (p < 0.001). Expression of Pla2g16 was then measured in control (db/+) and diabetic (db/db) mice that underwent unilateral femoral artery ligation. There was significantly reduced expression of Pla2g16 in the ischemic leg of both control and diabetic mice (by 51%), with significantly greater magnitude of reduction in the diabetic mice (by 79%). We conclude that AdPLA is downregulated in humans with PAD and in mice with hindlimb ischemia. Reduced AdPLA may contribute to impaired walking capacity in patients with PAD via its effects on skeletal muscle metabolism. Further studies are needed to fully characterize the role of AdPLA in PAD and to investigate its potential as a therapeutic target for alleviating symptoms of claudication.
Collapse
Affiliation(s)
- Caitlin Parmer
- Department of Medicine, Stanford University Medical Center, Palo Alto, CA, USA
| | | | - Henry S Cheng
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Grace Daher
- Research Division, Joslin Diabetes Center, and Harvard Medical School, Boston, MA, USA
| | - Alison Burkart
- Research Division, Joslin Diabetes Center, and Harvard Medical School, Boston, MA, USA
| | - Jonathan M Dreyfuss
- Research Division, Joslin Diabetes Center, and Harvard Medical School, Boston, MA, USA
| | - Hui Pan
- Research Division, Joslin Diabetes Center, and Harvard Medical School, Boston, MA, USA
| | - Joshua C Prenner
- Department of Medicine, McGaw Medical Center of Northwestern University, Chicago, IL, USA
| | | | - Reena Pande
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Stanislav Henkin
- Heart and Vascular Center, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Mark W Feinberg
- Cardiovascular Division, Brigham and Women's Hospital, and Harvard Medical School, Boston, MA, USA
| | - Mary Elizabeth Patti
- Research Division, Joslin Diabetes Center, and Harvard Medical School, Boston, MA, USA
| | - Mark A Creager
- Heart and Vascular Center, Dartmouth-Hitchcock Medical Center and Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| |
Collapse
|
26
|
Fan Z, Li N, Xu Z, Wu J, Fan X, Xu Y. An interaction between MKL1, BRG1, and C/EBPβ mediates palmitate induced CRP transcription in hepatocytes. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:194412. [PMID: 31356989 DOI: 10.1016/j.bbagrm.2019.194412] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/22/2019] [Accepted: 07/23/2019] [Indexed: 12/11/2022]
Abstract
Non-alcoholic steatohepatitis (NASH) is one of the most predominant disorders in metabolic syndrome. Induction of pro-inflammatory mediators in hepatocytes exposed to free fatty acids represents a hallmark event during NASH pathogenesis. C-reactive protein (CRP) is a prototypical pro-inflammatory mediator. In the present study, we investigated the mechanism by which megakaryocytic leukemia 1 (MKL1) mediates palmitate (PA) induced CRP transcription in hepatocytes. We report that over-expression of MKL1, but not MKL2, activated the CRP promoter whereas depletion or inhibition of MKL1 repressed the CRP promoter. MKL1 potentiated the induction of the CRP promoter activity by PA treatment. Importantly, MKL1 knockdown by siRNA or pharmaceutical inhibition by CCG-1423 attenuated the induction of endogenous CRP expression in hepatocytes. Similarly, primary hepatocytes isolated from wild type (WT) mice produced more CRP than those isolated from MKL1 deficient (KO) mice when stimulated with PA. Mechanistically, the sequence-specific transcription factor CCAAT-enhancer-binding protein (C/EBPβ) interacted with MKL1 and recruited MKL1 to activate CRP transcription. Reciprocally, MKL1 modulated C/EBPβ activity by recruiting the chromatin remodeling protein BRG1 to the CRP promoter to alter histone modifications. In conclusion, our data delineate a novel epigenetic mechanism underlying augmented hepatic inflammation during NASH pathogenesis.
Collapse
Affiliation(s)
- Zhiwen Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Nan Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Zheng Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Jiahao Wu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China
| | - Xiangshan Fan
- Department of Pathology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing, China.
| | - Yong Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Translational Medicine, Department of Pathophysiology, Nanjing Medical University, Nanjing, China; Institute of Biomedical Research, Liaocheng University, Liaocheng, China.
| |
Collapse
|
27
|
Rojo C, Zhang Q, Keleş S. iFunMed: Integrative functional mediation analysis of GWAS and eQTL studies. Genet Epidemiol 2019; 43:742-760. [PMID: 31328826 DOI: 10.1002/gepi.22217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/17/2019] [Accepted: 05/07/2019] [Indexed: 11/08/2022]
Abstract
Genome-wide association studies (GWAS) have successfully identified thousands of genetic variants contributing to disease and other phenotypes. However, significant obstacles hamper our ability to elucidate causal variants, identify genes affected by causal variants, and characterize the mechanisms by which genotypes influence phenotypes. The increasing availability of genome-wide functional annotation data is providing unique opportunities to incorporate prior information into the analysis of GWAS to better understand the impact of variants on disease etiology. Although there have been many advances in incorporating prior information into prioritization of trait-associated variants in GWAS, functional annotation data have played a secondary role in the joint analysis of GWAS and molecular (i.e., expression) quantitative trait loci (eQTL) data in assessing evidence for association. To address this, we develop a novel mediation framework, iFunMed, to integrate GWAS and eQTL data with the utilization of publicly available functional annotation data. iFunMed extends the scope of standard mediation analysis by incorporating information from multiple genetic variants at a time and leveraging variant-level summary statistics. Data-driven computational experiments convey how informative annotations improve single-nucleotide polymorphism (SNP) selection performance while emphasizing robustness of iFunMed to noninformative annotations. Application to Framingham Heart Study data indicates that iFunMed is able to boost detection of SNPs with mediation effects that can be attributed to regulatory mechanisms.
Collapse
Affiliation(s)
- Constanza Rojo
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Qi Zhang
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, Nebraska
| | - Sündüz Keleş
- Department of Statistics, University of Wisconsin-Madison, Madison, Wisconsin.,Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
28
|
Li L, Pan Z, Yang X. Key genes and co-expression network analysis in the livers of type 2 diabetes patients. J Diabetes Investig 2019; 10:951-962. [PMID: 30592156 PMCID: PMC6626963 DOI: 10.1111/jdi.12998] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 12/03/2018] [Accepted: 12/25/2018] [Indexed: 12/17/2022] Open
Abstract
AIMS/INTRODUCTION The incidence of type 2 diabetes is increasing worldwide. Hepatic insulin resistance and liver lipid accumulation contributes to type 2 diabetes development. The aim of the present study was to investigate the key gene pathways and co-expression networks in the livers of type 2 diabetes patients. MATERIALS AND METHODS Dataset GSE15653 containing nine healthy individuals and nine type 2 diabetes patients was downloaded from the National Center for Biotechnology Information Gene Expression Omnibus database. Differentially expressed genes were obtained from the livers of type 2 diabetes patients, annotated pathway enrichment and protein-protein interaction network analysis. Next, functional modules and transcription factor networks were constructed. Gene co-expression networks were analyzed by weighted correlation network analysis to identify key modules related to clinical traits, and the candidate key genes were validated in hepatic insulin resistance models in vitro. RESULTS A total of 778 differentially expressed genes were filtered in the livers of type 2 diabetes patients, pathway enrichment analysis identified ke y pathways, such as the mitogen-activated protein kinase signaling pathway, Hippo signaling pathway and hypoxia-inducible factor-1 signaling pathway, that were associated with type 2 diabetes. Several transcription factors of three functional modules identified from protein-protein interaction networks are likely to be implicated in type 2 diabetes. Furthermore, weighted correlation network analysis identified five modules that were shown to be highly correlated with type 2 diabetes and other clinical traits. Functional annotation showed that these modules were mainly enriched in pathways such as metabolic pathways, phosphoinositide 3-kinase-protein kinase B signaling pathway and natural killer cell-mediated cytotoxicity. UBE2M and GPER were upregulated in L02 and HepG2 models, whereas P2RY11 only upregulated in L02 model, and UBE2N only downregulated in HepG2 model at a significant level. CONCLUSIONS These results would offer new insights into hepatic insulin resistance, type 2 diabetes pathogenesis, development and drug discovery.
Collapse
Affiliation(s)
- Lu Li
- Department of PharmacyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| | - Zongfu Pan
- Department of PharmacyZhejiang Cancer HospitalHangzhouChina
| | - Xi Yang
- Department of PharmacyThe First Affiliated HospitalCollege of MedicineZhejiang UniversityHangzhouChina
| |
Collapse
|
29
|
A non-autonomous role of MKL1 in the activation of hepatic stellate cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2019; 1862:609-618. [DOI: 10.1016/j.bbagrm.2019.03.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 01/07/2019] [Accepted: 03/30/2019] [Indexed: 01/20/2023]
|
30
|
Kahl DJ, Hutchings KM, Lisabeth EM, Haak AJ, Leipprandt JR, Dexheimer T, Khanna D, Tsou PS, Campbell PL, Fox DA, Wen B, Sun D, Bailie M, Neubig RR, Larsen SD. 5-Aryl-1,3,4-oxadiazol-2-ylthioalkanoic Acids: A Highly Potent New Class of Inhibitors of Rho/Myocardin-Related Transcription Factor (MRTF)/Serum Response Factor (SRF)-Mediated Gene Transcription as Potential Antifibrotic Agents for Scleroderma. J Med Chem 2019; 62:4350-4369. [PMID: 30951312 PMCID: PMC6590913 DOI: 10.1021/acs.jmedchem.8b01772] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Through a phenotypic high-throughput screen using a serum response element luciferase promoter, we identified a novel 5-aryl-1,3,4-oxadiazol-2-ylthiopropionic acid lead inhibitor of Rho/myocardin-related transcription factor (MRTF)/serum response factor (SRF)-mediated gene transcription with good potency (IC50 = 180 nM). We were able to rapidly improve the cellular potency by 5 orders of magnitude guided by sharply defined and synergistic SAR. The remarkable potency and depth of the SAR, as well as the relatively low molecular weight of the series, suggests, but does not prove, that binding to the unknown molecular target may be occurring through a covalent mechanism. The series nevertheless has no observable cytotoxicity up to 100 μM. Ensuing pharmacokinetic optimization resulted in the development of two potent and orally bioavailable anti-fibrotic agents that were capable of dose-dependently reducing connective tissue growth factor gene expression in vitro as well as significantly reducing the development of bleomycin-induced dermal fibrosis in mice in vivo.
Collapse
Affiliation(s)
| | | | - Erika Mathes Lisabeth
- Department of Pharmacology and Toxicology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Andrew J Haak
- Department of Pharmacology and Toxicology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Jeffrey R Leipprandt
- Department of Pharmacology and Toxicology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Thomas Dexheimer
- Department of Pharmacology and Toxicology , Michigan State University , East Lansing , Michigan 48824 , United States
| | - Dinesh Khanna
- Department of Internal Medicine, Division of Rheumatology and Clinical Autoimmunity Center of Excellence , University of Michigan Medical Center , Ann Arbor , Michigan 48109 , United States
| | - Pei-Suen Tsou
- Department of Internal Medicine, Division of Rheumatology and Clinical Autoimmunity Center of Excellence , University of Michigan Medical Center , Ann Arbor , Michigan 48109 , United States
| | - Phillip L Campbell
- Department of Internal Medicine, Division of Rheumatology and Clinical Autoimmunity Center of Excellence , University of Michigan Medical Center , Ann Arbor , Michigan 48109 , United States
| | - David A Fox
- Department of Internal Medicine, Division of Rheumatology and Clinical Autoimmunity Center of Excellence , University of Michigan Medical Center , Ann Arbor , Michigan 48109 , United States
| | | | | | - Marc Bailie
- Michigan State University in Vivo Facility , East Lansing , Michigan 48824 , United States
| | - Richard R Neubig
- Department of Pharmacology and Toxicology , Michigan State University , East Lansing , Michigan 48824 , United States
| | | |
Collapse
|
31
|
Lisabeth EM, Kahl D, Gopallawa I, Haynes SE, Misek SA, Campbell PL, Dexheimer TS, Khanna D, Fox DA, Jin X, Martin BR, Larsen SD, Neubig RR. Identification of Pirin as a Molecular Target of the CCG-1423/CCG-203971 Series of Antifibrotic and Antimetastatic Compounds. ACS Pharmacol Transl Sci 2019; 2:92-100. [PMID: 32039344 DOI: 10.1021/acsptsci.8b00048] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
A series of compounds (including CCG-1423 and CCG-203971) discovered through an MRTF/SRF-dependent luciferase screen has shown remarkable efficacy in a variety of in vitro and in vivo models, including significant reduction of melanoma metastasis and bleomycin- induced fibrosis. Although these compounds are efficacious in these disease models, the molecular target is unknown. Here, we describe affinity isolation-based target identification efforts which yielded pirin, an iron-dependent cotranscription factor, as a target of this series of compounds. Using biophysical techniques including isothermal titration calorimetry and X-ray crystallography, we verify that pirin binds these compounds in vitro. We also show with genetic approaches that pirin modulates MRTF- dependent luciferase reporter activity. Finally, using both siRNA and a previously validated pirin inhibitor, we show a role for pirin in TGF-β- induced gene expression in primary dermal fibroblasts. A recently developed analog, CCG-257081, which co crystallizes with pirin, is also effective in the prevention of bleomycin-induced dermal fibrosis.
Collapse
Affiliation(s)
- Erika M Lisabeth
- Department of Pharmacology & Toxicology and Michigan State University, East Lansing, Michigan, 48824, United States
| | - Dylan Kahl
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Indiwari Gopallawa
- Department of Pharmacology & Toxicology and Michigan State University, East Lansing, Michigan, 48824, United States
| | - Sarah E Haynes
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Sean A Misek
- Department of Pharmacology & Toxicology and Michigan State University, East Lansing, Michigan, 48824, United States
| | - Phillip L Campbell
- Department of Internal Medicine, Division of Rheumatology, and University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Thomas S Dexheimer
- Department of Pharmacology & Toxicology and Michigan State University, East Lansing, Michigan, 48824, United States
| | - Dinesh Khanna
- Department of Internal Medicine, Division of Rheumatology, and University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - David A Fox
- Department of Internal Medicine, Division of Rheumatology, and University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Xiangshu Jin
- Department of Biochemistry, Michigan State University, East Lansing, Michigan, 48824, United States
| | - Brent R Martin
- Department of Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Scott D Larsen
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, Michigan, 48109, United States.,Vahlteich Medicinal Chemistry Core, College of Pharmacy, University of Michigan, Ann Arbor, Michigan, 48109, United States
| | - Richard R Neubig
- Department of Pharmacology & Toxicology and Michigan State University, East Lansing, Michigan, 48824, United States
| |
Collapse
|
32
|
Osataphan S, Macchi C, Singhal G, Chimene-Weiss J, Sales V, Kozuka C, Dreyfuss JM, Pan H, Tangcharoenpaisan Y, Morningstar J, Gerszten R, Patti ME. SGLT2 inhibition reprograms systemic metabolism via FGF21-dependent and -independent mechanisms. JCI Insight 2019; 4:123130. [PMID: 30843877 DOI: 10.1172/jci.insight.123130] [Citation(s) in RCA: 139] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 01/17/2019] [Indexed: 12/19/2022] Open
Abstract
Pharmacologic inhibition of the renal sodium/glucose cotransporter-2 induces glycosuria and reduces glycemia. Given that SGLT2 inhibitors (SGLT2i) reduce mortality and cardiovascular risk in type 2 diabetes, improved understanding of molecular mechanisms mediating these metabolic effects is required. Treatment of obese but nondiabetic mice with the SGLT2i canagliflozin (CANA) reduces adiposity, improves glucose tolerance despite reduced plasma insulin, increases plasma ketones, and improves plasma lipid profiles. Utilizing an integrated transcriptomic-metabolomics approach, we demonstrate that CANA modulates key nutrient-sensing pathways, with activation of 5' AMP-activated protein kinase (AMPK) and inhibition of mechanistic target of rapamycin (mTOR), independent of insulin or glucagon sensitivity or signaling. Moreover, CANA induces transcriptional reprogramming to activate catabolic pathways, increase fatty acid oxidation, reduce hepatic steatosis and diacylglycerol content, and increase hepatic and plasma levels of FGF21. Given that these phenotypes mirror the effects of FGF21 to promote lipid oxidation, ketogenesis, and reduction in adiposity, we hypothesized that FGF21 is required for CANA action. Using FGF21-null mice, we demonstrate that FGF21 is not required for SGLT2i-mediated induction of lipid oxidation and ketogenesis but is required for reduction in fat mass and activation of lipolysis. Taken together, these data demonstrate that SGLT2 inhibition triggers a fasting-like transcriptional and metabolic paradigm but requires FGF21 for reduction in adiposity.
Collapse
Affiliation(s)
- Soravis Osataphan
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA.,Department of Pathology, Srinakharinwirot University, Bangkok, Thailand
| | - Chiara Macchi
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Garima Singhal
- Harvard Medical School, Boston, Massachusetts, USA.,Division of Endocrinology and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Jeremy Chimene-Weiss
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Vicencia Sales
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Chisayo Kozuka
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| | - Jonathan M Dreyfuss
- Harvard Medical School, Boston, Massachusetts, USA.,Bioinformatics and Biostatistics Core, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Hui Pan
- Harvard Medical School, Boston, Massachusetts, USA.,Bioinformatics and Biostatistics Core, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Yanin Tangcharoenpaisan
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA
| | - Jordan Morningstar
- Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Robert Gerszten
- Harvard Medical School, Boston, Massachusetts, USA.,Division of Cardiovascular Medicine, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Mary-Elizabeth Patti
- Section of Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Boston, Massachusetts, USA.,Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
33
|
Gupta MK, Vadde R. Identification and characterization of differentially expressed genes in Type 2 Diabetes using in silico approach. Comput Biol Chem 2019; 79:24-35. [PMID: 30708140 DOI: 10.1016/j.compbiolchem.2019.01.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 12/26/2018] [Accepted: 01/23/2019] [Indexed: 12/14/2022]
Abstract
Diabetes mellitus is clinically characterized by hyperglycemia. Though many studies have been done to understand the mechanism of Type 2 Diabetes (T2D), however, the complete network of diabetes and its associated disorders through polygenic involvement is still under debate. The present study designed to re-analyze publicly available T2D related microarray raw datasets present in GEO database and T2D genes information present in GWAS catalog for screening out differentially expressed genes (DEGs) and identify key hub genes associated with T2D. T2D related microarray data downloaded from Gene Expression Omnibus (GEO) database and re-analysis performed with in house R packages scripts for background correction, normalization and identification of DEGs in T2D. Also retrieved T2D related DEGs information from GWAS catalog. Both DEGs lists were grouped after removal of overlapping genes. These screened DEGs were utilized further for identification and characterization of key hub genes in T2D and its associated diseases using STRING, WebGestalt and Panther databases. Computational analysis reveal that out of 99 identified key hub gene candidates from 348 DEGs, only four genes (CCL2, ELMO1, VEGFA and TCF7L2) along with FOS playing key role in causing T2D and its associated disorders, like nephropathy, neuropathy, rheumatoid arthritis and cancer via p53 or Wnt signaling pathways. MIR-29, and MAZ_Q6 are identified potential target microRNA and TF along with probable drugs alprostadil, collagenase and dinoprostone for the key hub gene candidates. The results suggest that identified key DEGs may play promising roles in prevention of diabetes.
Collapse
Affiliation(s)
- Manoj Kumar Gupta
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa 516003, Andhra Pradesh, India.
| | - Ramakrishna Vadde
- Department of Biotechnology & Bioinformatics, Yogi Vemana University, Kadapa 516003, Andhra Pradesh, India.
| |
Collapse
|
34
|
Megakaryocytic leukemia 1 (MKL1) mediates high glucose induced epithelial-mesenchymal transition by activating LOX transcription. Biochem Biophys Res Commun 2018; 509:633-640. [PMID: 30553442 DOI: 10.1016/j.bbrc.2018.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 12/04/2018] [Indexed: 12/17/2022]
Abstract
Diabetic retinopathy (DR) is one of the most devastating complications of diabetes mellitus. When exposed to high glucose (HG), retinal epithelial cells undergo profound alterations both morphologically and functionally in a well-conserved process known as epithelial-to-mesenchymal transition (EMT). The mechanism governing HG-induced EMT in retinal epithelial cells is not completely understood. Here we report that treatment with 25 mM glucose led to EMT in retinal pigmented epithelial cells (RPE) characterized by a simultaneous down-regulation of E-Cadherin (encoded by CDH1) and up-regulation of alpha smooth muscle actin (encoded by ACTA2). HG-induced EMT in RPEs was accompanied by augmented expression and enhanced nuclear enrichment of MKL1, a transcriptional modulator. In contrast, MKL1 knockdown by siRNA or inhibition by CCG-1423 abrogated HG-induced EMT in RPEs. Of interest, MKL1 mediated the transcriptional activation of LOX, a mesenchymal marker, in RPEs in response to HG stimulation. Mechanistically, MKL1 interacted with and was recruited by AP-1 to the proximal LOX promoter to promote LOX trans-activation likely through altering the chromatin structure. Finally, LOX depletion by siRNA or inhibition by aminopropionitrile in RPEs abolished HG-induced EMT. In conclusion, our data support a role for MKL1 in mediating HG-induced EMT in retinal epithelial cells via epigenetic activation of LOX transcription.
Collapse
|
35
|
Saxena A, Sachin K. A Network Biology Approach for Assessing the Role of Pathologic Adipose Tissues in Insulin Resistance Using Meta-analysis of Microarray Datasets. Curr Genomics 2018; 19:630-666. [PMID: 30386174 PMCID: PMC6194434 DOI: 10.2174/1389202919666180726125645] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 05/10/2018] [Accepted: 07/16/2018] [Indexed: 11/22/2022] Open
Abstract
Background The role of adipose tissue in Insulin resistance (IR) and Type 2 Diabetes (T2D) has well been received in the biomedical community; being a precursor of T2D, identification of the molecular basis of IR is therefore, vital to elucidate T2D- pathogenesis and meta-analysis of previously conducted microarray studies provides an inexpensive approach to achieve this end. Methods In this study, we have carried out a statistical meta-analysis of 157 microarray datasets from five independent studies and identified a meta-signature of 1,511 genes; their functional meaning was elucidated by integrated pathways-analysis. Further, a protein-protein interaction network was constructed and key genes along with their high confidence transcriptional- and epigenetic-mediators were identified using a network biology approach. Results Various inflammation- and immune system-related pathways such as TGF-β signaling, IL7 signaling, Neutrophil degranulation, and Chemokine signaling etc. were enriched in sick adipose tissues; identified transcription factors, and microRNAs were also found to regulate processes relevant to IR/T2D pathophysiology. Conclusion This study endorses the development of effective bioinformatics workflow and further grants an indication for the acceptance of adiposopathy as the root mechanistic pathology that poses risk for development of type 2 diabetes; concept of adipospathy in place of metabolic syndrome will open the possibility to design drugs, those will ameliorate adipose functions and hence proved to be more effective against Type 2 Diabetes.
Collapse
Affiliation(s)
- Aditya Saxena
- 1Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura (U.P.), India; 2Uttarakhand Technical University, Dehradun (U.K.), India; 3Department of Biochemistry and Biotechnology, S.B.S. (PG) Institute of Biomedical Sciences & Research, Dehradun (U.K.), India
| | - Kumar Sachin
- 1Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura (U.P.), India; 2Uttarakhand Technical University, Dehradun (U.K.), India; 3Department of Biochemistry and Biotechnology, S.B.S. (PG) Institute of Biomedical Sciences & Research, Dehradun (U.K.), India
| |
Collapse
|
36
|
Timmons JA, Atherton PJ, Larsson O, Sood S, Blokhin IO, Brogan RJ, Volmar CH, Josse AR, Slentz C, Wahlestedt C, Phillips SM, Phillips BE, Gallagher IJ, Kraus WE. A coding and non-coding transcriptomic perspective on the genomics of human metabolic disease. Nucleic Acids Res 2018; 46:7772-7792. [PMID: 29986096 PMCID: PMC6125682 DOI: 10.1093/nar/gky570] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 05/23/2018] [Accepted: 06/13/2018] [Indexed: 12/13/2022] Open
Abstract
Genome-wide association studies (GWAS), relying on hundreds of thousands of individuals, have revealed >200 genomic loci linked to metabolic disease (MD). Loss of insulin sensitivity (IS) is a key component of MD and we hypothesized that discovery of a robust IS transcriptome would help reveal the underlying genomic structure of MD. Using 1,012 human skeletal muscle samples, detailed physiology and a tissue-optimized approach for the quantification of coding (>18,000) and non-coding (>15,000) RNA (ncRNA), we identified 332 fasting IS-related genes (CORE-IS). Over 200 had a proven role in the biochemistry of insulin and/or metabolism or were located at GWAS MD loci. Over 50% of the CORE-IS genes responded to clinical treatment; 16 quantitatively tracking changes in IS across four independent studies (P = 0.0000053: negatively: AGL, G0S2, KPNA2, PGM2, RND3 and TSPAN9 and positively: ALDH6A1, DHTKD1, ECHDC3, MCCC1, OARD1, PCYT2, PRRX1, SGCG, SLC43A1 and SMIM8). A network of ncRNA positively related to IS and interacted with RNA coding for viral response proteins (P < 1 × 10-48), while reduced amino acid catabolic gene expression occurred without a change in expression of oxidative-phosphorylation genes. We illustrate that combining in-depth physiological phenotyping with robust RNA profiling methods, identifies molecular networks which are highly consistent with the genetics and biochemistry of human metabolic disease.
Collapse
Affiliation(s)
- James A Timmons
- Division of Genetics and Molecular Medicine, King's College London, London, UK
- Scion House, Stirling University Innovation Park, Stirling, UK
| | | | - Ola Larsson
- Department of Oncology-Pathology, Science For Life Laboratory, Stockholm, Sweden
| | - Sanjana Sood
- Division of Genetics and Molecular Medicine, King's College London, London, UK
| | | | - Robert J Brogan
- Scion House, Stirling University Innovation Park, Stirling, UK
| | | | | | - Cris Slentz
- Duke University School of Medicine, Durham, USA
| | - Claes Wahlestedt
- Department of Oncology-Pathology, Science For Life Laboratory, Stockholm, Sweden
| | | | | | - Iain J Gallagher
- Scion House, Stirling University Innovation Park, Stirling, UK
- School of Health Sciences and Sport, University of Stirling, Stirling, UK
| | | |
Collapse
|
37
|
Jung S. Implications of publicly available genomic data resources in searching for therapeutic targets of obesity and type 2 diabetes. Exp Mol Med 2018; 50:1-13. [PMID: 29674722 PMCID: PMC5938056 DOI: 10.1038/s12276-018-0066-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2017] [Accepted: 01/28/2018] [Indexed: 12/29/2022] Open
Abstract
Obesity and type 2 diabetes (T2D) are two major conditions that are related to metabolic disorders and affect a large population. Although there have been significant efforts to identify their therapeutic targets, few benefits have come from comprehensive molecular profiling. This limited availability of comprehensive molecular profiling of obesity and T2D may be due to multiple challenges, as these conditions involve multiple organs and collecting tissue samples from subjects is more difficult in obesity and T2D than in other diseases, where surgical treatments are popular choices. While there is no repository of comprehensive molecular profiling data for obesity and T2D, multiple existing data resources can be utilized to cover various aspects of these conditions. This review presents studies with available genomic data resources for obesity and T2D and discusses genome-wide association studies (GWAS), a knockout (KO)-based phenotyping study, and gene expression profiles. These studies, based on their assessed coverage and characteristics, can provide insights into how such data can be utilized to identify therapeutic targets for obesity and T2D.
Collapse
Affiliation(s)
- Sungwon Jung
- Department of Genome Medicine and Science, Gachon University School of Medicine, Incheon, Republic of Korea. .,Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Incheon, Republic of Korea.
| |
Collapse
|
38
|
Srf destabilizes cellular identity by suppressing cell-type-specific gene expression programs. Nat Commun 2018; 9:1387. [PMID: 29643333 PMCID: PMC5895821 DOI: 10.1038/s41467-018-03748-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Accepted: 03/09/2018] [Indexed: 01/07/2023] Open
Abstract
Multicellular organisms consist of multiple cell types. The identity of these cells is primarily maintained by cell-type-specific gene expression programs; however, mechanisms that suppress these programs are poorly defined. Here we show that serum response factor (Srf), a transcription factor that is activated by various extracellular stimuli, can repress cell-type-specific genes and promote cellular reprogramming to pluripotency. Manipulations that decrease β-actin monomer quantity result in the nuclear accumulation of Mkl1 and the activation of Srf, which downregulate cell-type-specific genes and alter the epigenetics of regulatory regions and chromatin organization. Mice overexpressing Srf exhibit various pathologies including an ulcerative colitis-like symptom and a metaplasia-like phenotype in the pancreas. Our results demonstrate an unexpected function of Srf via a mechanism by which extracellular stimuli actively destabilize cell identity and suggest Srf involvement in a wide range of diseases. The transcription factor Srf is a central regulator of immediate-early and actin cytoskeletal genes. Here the authors show that Srf is activated by repression of β-actin, promoting iPSC reprogramming of neural progenitor cells and hepatoblasts by repressing cell-type specific genes.
Collapse
|
39
|
Wu C, Chen X, Shu J, Lee CT. Whole-genome expression analyses of type 2 diabetes in human skin reveal altered immune function and burden of infection. Oncotarget 2018; 8:34601-34609. [PMID: 28427244 PMCID: PMC5470994 DOI: 10.18632/oncotarget.16118] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/20/2017] [Indexed: 12/26/2022] Open
Abstract
Skin disorders are among most common complications associated with type 2 diabetes mellitus (T2DM). Although T2DM patients are known to have increased risk of infections and other T2DM-related skin disorders, their molecular mechanisms are largely unknown. This study aims to identify dysregulated genes and gene networks that are associated with T2DM in human skin. We compared the expression profiles of 56,318 transcribed genes on 74 T2DM cases and 148 gender- age-, and race-matched non-diabetes controls from the Genotype-Tissue Expression (GTEx) database. RNA-Sequencing data indicates that diabetic skin is characterized by increased expression of genes that are related to immune responses (CCL20, CXCL9, CXCL10, CXCL11, CXCL13, and CCL18), JAK/STAT signaling pathway (JAK3, STAT1, and STAT2), tumor necrosis factor superfamily (TNFSF10 and TNFSF15), and infectious disease pathways (OAS1, OAS2, OAS3, and IFIH1). Genes in cell adhesion molecules pathway (NCAM1 and L1CAM) and collagen family (PCOLCE2 and COL9A3) are downregulated, suggesting structural changes in the skin of T2DM. For the first time, to the best of our knowledge, this pioneer analytic study reports comprehensive unbiased gene expression changes and dysregulated pathways in the non-diseased skin of T2DM patients. This comprehensive understanding derived from whole-genome expression profiles could advance our knowledge in determining molecular targets for the prevention and treatment of T2DM-associated skin disorders.
Collapse
Affiliation(s)
- Chun Wu
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, FL, USA
| | - Xiaopan Chen
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, P.R. China
| | - Jing Shu
- Department of Reproductive Endocrinology, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, P.R. China
| | - Chun-Ting Lee
- Department of Neurology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
40
|
Singhal A, Agrawal A, Ling J. Regulation of insulin resistance and type II diabetes by hepatitis C virus infection: A driver function of circulating miRNAs. J Cell Mol Med 2018; 22:2071-2085. [PMID: 29411512 PMCID: PMC5867149 DOI: 10.1111/jcmm.13553] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 01/04/2018] [Indexed: 12/15/2022] Open
Abstract
Hepatitis C virus (HCV) infection is a serious worldwide healthcare issue. Its association with various liver diseases including hepatocellular carcinoma (HCC) is well studied. However, the study on the relationship between HCV infection and the development of insulin resistance and diabetes is very limited. Current research has already elucidated some underlying mechanisms, especially on the regulation of metabolism and insulin signalling by viral proteins. More studies have emerged recently on the correlation between HCV infection‐derived miRNAs and diabetes and insulin resistance. However, no studies have been carried out to directly address if these miRNAs, especially circulating miRNAs, have causal effects on the development of insulin resistance and diabetes. Here, we proposed a new perspective that circulating miRNAs can perform regulatory functions to modulate gene expression in peripheral tissues leading to insulin resistance and diabetes, rather than just a passive factor associated with these pathological processes. The detailed rationales were elaborated through comprehensive literature review and bioinformatic analyses. miR‐122 was identified to be one of the most potential circulating miRNAs to cause insulin resistance. This result along with the idea about the driver function of circulating miRNAs will promote further investigations that eventually lead to the development of novel strategies to treat HCV infection‐associated extrahepatic comorbidities.
Collapse
Affiliation(s)
- Adit Singhal
- Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| | | | - Jun Ling
- Geisinger Commonwealth School of Medicine, Scranton, PA, USA
| |
Collapse
|
41
|
Giorgetti E, Yu Z, Chua JP, Shimamura R, Zhao L, Zhu F, Venneti S, Pennuto M, Guan Y, Hung G, Lieberman AP. Rescue of Metabolic Alterations in AR113Q Skeletal Muscle by Peripheral Androgen Receptor Gene Silencing. Cell Rep 2017; 17:125-136. [PMID: 27681426 DOI: 10.1016/j.celrep.2016.08.084] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 07/05/2016] [Accepted: 08/24/2016] [Indexed: 01/09/2023] Open
Abstract
Spinal and bulbar muscular atrophy (SBMA), a progressive degenerative disorder, is caused by a CAG/glutamine expansion in the androgen receptor (polyQ AR). Recent studies demonstrate that skeletal muscle is an important site of toxicity that contributes to the SBMA phenotype. Here, we sought to identify critical pathways altered in muscle that underlie disease manifestations in AR113Q mice. This led to the unanticipated identification of gene expression changes affecting regulators of carbohydrate metabolism, similar to those triggered by denervation. AR113Q muscle exhibits diminished glycolysis, altered mitochondria, and an impaired response to exercise. Strikingly, the expression of genes regulating muscle energy metabolism is rescued following peripheral polyQ AR gene silencing by antisense oligonucleotides (ASO), a therapeutic strategy that alleviates disease. Our data establish the occurrence of a metabolic imbalance in SBMA muscle triggered by peripheral expression of the polyQ AR and indicate that alterations in energy utilization contribute to non-neuronal disease manifestations.
Collapse
Affiliation(s)
- Elisa Giorgetti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Zhigang Yu
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jason P Chua
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Ryosuke Shimamura
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Fan Zhu
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Sriram Venneti
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Maria Pennuto
- Dulbecco Telethon Institute, Centre for Integrative Biology, University of Trento, Trento 38123, Italy
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Gene Hung
- Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Andrew P Lieberman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
42
|
Kikuchi K, Shiota J, Yamada T, Ishikawa M, Ihara D, Fukuchi M, Tsuda M, Tabuchi A. Rho signaling inhibitor, CCG-1423, inhibits axonal elongation and dendritic complexity of rat cortical neurons. Biochem Biophys Res Commun 2017; 492:474-479. [DOI: 10.1016/j.bbrc.2017.08.071] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/18/2017] [Indexed: 10/19/2022]
|
43
|
Ye G, Huang K, Yu J, Zhao L, Zhu X, Yang Q, Li W, Jiang Y, Zhuang B, Liu H, Shen Z, Wang D, Yan L, Zhang L, Zhou H, Hu Y, Deng H, Liu H, Li G, Qi X. MicroRNA-647 Targets SRF-MYH9 Axis to Suppress Invasion and Metastasis of Gastric Cancer. Theranostics 2017; 7:3338-3353. [PMID: 28900514 PMCID: PMC5595136 DOI: 10.7150/thno.20512] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 05/29/2017] [Indexed: 12/28/2022] Open
Abstract
MicroRNAs (miRNAs) play important roles in regulating tumour development and progression. Here we show that miR-647 is repressed in gastric cancer (GC), and associated with GC metastasis. Moreover, we identify that miR-647 can suppress GC cell migration and invasion in vitro. Mechanistically, we confirm miR-647 directly binds to the 3' untranslated regions of SRF mRNA, and SRF binds to the CArG box located at the MYH9 promoter. CCG-1423, an inhibitor of RhoA/SRF-mediated gene transcription, inhibits the expression of MYH9, especially in SRF downregulated cells. Overexpression of miR-647 inhibits MGC 80-3 cells' metastasis in orthotropic GC models, but increasing SRF expression in these cells reverses this change. Importantly, we found the synergistic inhibition effect of CCG-1423 and agomir-647, an engineered miRNA mimic, on cancer metastasis in orthotropic GC models. Our study demonstrates that miR-647 functions as a tumor metastasis suppressor in GC by targeting SRF/MYH9 axis.
Collapse
Affiliation(s)
- Gengtai Ye
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Kunzhai Huang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Liying Zhao
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Xianjun Zhu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Qingbin Yang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Wende Li
- Guangdong Key Laboratory of Laboratory Animal, Guangdong Laboratory Animal Monitoring Institute, Guangzhou 510663, China
| | - Yuming Jiang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Baoxiong Zhuang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Hao Liu
- Leder Human Biology and Translational Medicine, Biology and Biomedical Sciences, Division of Medical Sciences, Harvard Medical School, Boston, MA 02115
| | - Zhiyong Shen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Da Wang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Li Yan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Lei Zhang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Haipeng Zhou
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Yanfeng Hu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Haijun Deng
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Hao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Guoxin Li
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| | - Xiaolong Qi
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, 510515 China
| |
Collapse
|
44
|
Abstract
Protein function can be regulated via post-translational modifications by numerous enzymatic and non-enzymatic mechanisms, including oxidation of cysteine and methionine residues. Redox-dependent regulatory mechanisms have been identified for nearly every cellular process, but the major paradigm has been that cellular components are oxidized (damaged) by reactive oxygen species (ROS) in a relatively unspecific way, and then reduced (repaired) by designated reductases. While this scheme may work with cysteine, it cannot be ascribed to other residues, such as methionine, whose reaction with ROS is too slow to be biologically relevant. However, methionine is clearly oxidized in vivo and enzymes for its stereoselective reduction are present in all three domains of life. Here, we revisit the chemistry and biology of methionine oxidation, with emphasis on its generation by enzymes from the monooxygenase family. Particular attention is placed on MICALs, a recently discovered family of proteins that harbor an unusual flavin-monooxygenase domain with an NADPH-dependent methionine sulfoxidase activity. Based on structural and kinetic information we provide a rational framework to explain MICAL mechanism, inhibition, and regulation. Methionine residues that are targeted by MICALs are reduced back by methionine sulfoxide reductases, suggesting that reversible methionine oxidation may be a general mechanism analogous to the regulation by phosphorylation by kinases/phosphatases. The identification of new enzymes that catalyze the oxidation of methionine will open a new area of research at the forefront of redox signaling.
Collapse
Affiliation(s)
- Bruno Manta
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Vadim N Gladyshev
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
45
|
Pharmacological intervention of MKL/SRF signaling by CCG-1423 impedes endothelial cell migration and angiogenesis. Angiogenesis 2017. [PMID: 28638990 DOI: 10.1007/s10456-017-9560-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
De novo synthesis of cytoskeleton-regulatory proteins triggered by the megakaryoblastic leukemia (MKL)/serum response factor (SRF) transcriptional system in response to pro-angiogenic growth factors lies at the heart of endothelial cell (EC) migration (a critical element of angiogenesis) and neovascularization. This study explores whether pharmacological intervention of MKL/SRF signaling axis by CCG-1423 is able to suppress angiogenesis. Our studies show that CCG-1423 inhibits migration and cord morphogenesis of EC in vitro and sprouting angiogenesis ex vivo and in vivo, suggesting CCG-1423 could be a novel anti-angiogenic agent. Kymography analyses of membrane dynamics of EC revealed that CCG-1423 treatment causes a major defect in membrane protrusion. CCG-1423 treatment led to attenuated expression of several actin-binding proteins that are important for driving membrane protrusion including ArpC2, VASP, and profilin1 (Pfn1) with the most drastic effect seen on the expression of Pfn1. Finally, depletion of Pfn1 alone is also sufficient for a dramatic decrease in sprouting angiogenesis of EC in vitro and ex vivo, further suggesting that Pfn1 depletion may be one of the mechanisms of the anti-angiogenic action of CCG-1423.
Collapse
|
46
|
Joy M, Gau D, Castellucci N, Prywes R, Roy P. The myocardin-related transcription factor MKL co-regulates the cellular levels of two profilin isoforms. J Biol Chem 2017; 292:11777-11791. [PMID: 28546428 DOI: 10.1074/jbc.m117.781104] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/24/2017] [Indexed: 12/26/2022] Open
Abstract
Megakaryoblastic leukemia (MKL)/serum-response factor (SRF)-mediated gene transcription is a highly conserved mechanism that connects dynamic reorganization of the actin cytoskeleton to regulation of expression of a wide range of genes, including SRF itself and many important structural and regulatory components of the actin cytoskeleton. In this study, we examined the possible role of MKL/SRF in the context of regulation of profilin (Pfn), a major controller of actin dynamics and actin cytoskeletal remodeling in cells. We demonstrated that despite being located on different genomic loci, two major isoforms of Pfn (Pfn1 and Pfn2) are co-regulated by a common mechanism involving the action of MKL that is independent of its SRF-related activity. We found that MKL co-regulates the expression of Pfn isoforms indirectly by modulating signal transducer and activator of transcription 1 (STAT1) and utilizing its SAP-domain function. Unexpectedly, our studies revealed that cellular externalization, rather than transcription of Pfn1, is affected by the perturbations of MKL. We further demonstrated that MKL can influence cell migration by modulating Pfn1 expression, indicating a functional connection between MKL and Pfn1 in actin-dependent cellular processes. Finally, we provide initial evidence supporting the ability of Pfn to influence MKL and SRF expression. Collectively, these findings suggest that Pfn may play a role in a possible feedback loop of the actin/MKL/SRF signaling circuit.
Collapse
Affiliation(s)
- Marion Joy
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - David Gau
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Nevin Castellucci
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219
| | - Ron Prywes
- Department of Biological Sciences, Columbia University, New York, New York 10027
| | - Partha Roy
- Departments of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania 15219; Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219; Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania 15219.
| |
Collapse
|
47
|
Tissue Non-Specific Genes and Pathways Associated with Diabetes: An Expression Meta-Analysis. Genes (Basel) 2017; 8:genes8010044. [PMID: 28117714 PMCID: PMC5295038 DOI: 10.3390/genes8010044] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/03/2017] [Accepted: 01/13/2017] [Indexed: 12/16/2022] Open
Abstract
We performed expression studies to identify tissue non-specific genes and pathways of diabetes by meta-analysis. We searched curated datasets of the Gene Expression Omnibus (GEO) database and identified 13 and five expression studies of diabetes and insulin responses at various tissues, respectively. We tested differential gene expression by empirical Bayes-based linear method and investigated gene set expression association by knowledge-based enrichment analysis. Meta-analysis by different methods was applied to identify tissue non-specific genes and gene sets. We also proposed pathway mapping analysis to infer functions of the identified gene sets, and correlation and independent analysis to evaluate expression association profile of genes and gene sets between studies and tissues. Our analysis showed that PGRMC1 and HADH genes were significant over diabetes studies, while IRS1 and MPST genes were significant over insulin response studies, and joint analysis showed that HADH and MPST genes were significant over all combined data sets. The pathway analysis identified six significant gene sets over all studies. The KEGG pathway mapping indicated that the significant gene sets are related to diabetes pathogenesis. The results also presented that 12.8% and 59.0% pairwise studies had significantly correlated expression association for genes and gene sets, respectively; moreover, 12.8% pairwise studies had independent expression association for genes, but no studies were observed significantly different for expression association of gene sets. Our analysis indicated that there are both tissue specific and non-specific genes and pathways associated with diabetes pathogenesis. Compared to the gene expression, pathway association tends to be tissue non-specific, and a common pathway influencing diabetes development is activated through different genes at different tissues.
Collapse
|
48
|
Gonzalez-Franquesa A, Patti ME. Insulin Resistance and Mitochondrial Dysfunction. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:465-520. [DOI: 10.1007/978-3-319-55330-6_25] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
Williams ED, Rogers SC, Zhang X, Azhar G, Wei JY. p49/STRAP, a Serum Response Factor Binding Protein (SRFBP1), Is Involved in the Redistribution of Cytoskeletal F-Actin Proteins during Glucose Deprivation. J Nutr Health Aging 2017; 21:1142-1150. [PMID: 29188873 DOI: 10.1007/s12603-017-0925-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The functional decline that usually accompanies adult aging also encompasses cellular changes including cytoplasmic architecture. In addition to their role in cytoskeletal structure, actin microfilaments have important roles in various cellular processes, including cell-to-cell communication and intracellular signaling. Age-related diseases and late-stage cellular morphological appearances often correlate with altered f-actin structure, which has been observed most notably in cancer. What remains less clear are the molecular pathways that may be involved in normal and premature aging-induced f-actin changes. We report herein that p49/STRAP, a serum response factor binding protein (SRFBP1), is increased with normal aging and appears to be sensitive to low glucose-exposure. Our study results suggest that increased levels of p49/STRAP expression tend to correlate with f-actin redistribution genes, particularly cofilin, while siRNA-mediated knockdown of p49/STRAP resulted in a reduction of thymosin-β4. Furthermore, with the redistribution of f-actin, we observed an increase in the intermediate filament vimentin, compatible with the notion that vimentin may be increased due to its greater role in cytoskeletal dynamics during advancing population doubling levels (PDLs) and in response to a low-glucose exposure. Taken together, these data suggest that p49/STRAP may play a role in glucose-deprivation associated cytoskeletal changes.
Collapse
Affiliation(s)
- E D Williams
- Jeanne Y. Wei, M.D., Ph.D, Reynolds Institute on Aging, Department of Geriatrics, University of Arkansas for Medical Sciences, 4301 W. Markham St. #748, Little Rock, AR 72205, USA, Phone: (501) 526-6800, Fax: (501) 686-5300,
| | | | | | | | | |
Collapse
|
50
|
Kumar S, Ambrosini G, Bucher P. SNP2TFBS - a database of regulatory SNPs affecting predicted transcription factor binding site affinity. Nucleic Acids Res 2016; 45:D139-D144. [PMID: 27899579 PMCID: PMC5210548 DOI: 10.1093/nar/gkw1064] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/05/2016] [Accepted: 10/24/2016] [Indexed: 01/21/2023] Open
Abstract
SNP2TFBS is a computational resource intended to support researchers investigating the molecular mechanisms underlying regulatory variation in the human genome. The database essentially consists of a collection of text files providing specific annotations for human single nucleotide polymorphisms (SNPs), namely whether they are predicted to abolish, create or change the affinity of one or several transcription factor (TF) binding sites. A SNP's effect on TF binding is estimated based on a position weight matrix (PWM) model for the binding specificity of the corresponding factor. These data files are regenerated at regular intervals by an automatic procedure that takes as input a reference genome, a comprehensive SNP catalogue and a collection of PWMs. SNP2TFBS is also accessible over a web interface, enabling users to view the information provided for an individual SNP, to extract SNPs based on various search criteria, to annotate uploaded sets of SNPs or to display statistics about the frequencies of binding sites affected by selected SNPs. Homepage: http://ccg.vital-it.ch/snp2tfbs/.
Collapse
Affiliation(s)
- Sunil Kumar
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland.,Swiss Institute of Bioinformatics (SIB), CH-1015 Lausanne, Switzerland
| | - Giovanna Ambrosini
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland.,Swiss Institute of Bioinformatics (SIB), CH-1015 Lausanne, Switzerland
| | - Philipp Bucher
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland .,Swiss Institute of Bioinformatics (SIB), CH-1015 Lausanne, Switzerland
| |
Collapse
|