1
|
Li X, Hallajzadeh J. Circulating microRNAs and physical activity: Impact in diabetes. Clin Chim Acta 2025; 569:120178. [PMID: 39900127 DOI: 10.1016/j.cca.2025.120178] [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: 12/26/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
The term "ci-miRNAs," or "circulating microRNAs," refers to extracellular microRNAs (miRNAs) that exist outside of cells and can be detected in various bodily fluids, including blood, saliva, urine, and breast milk. These ci-miRNAs play a role in regulating gene expression and are mainly recognized for their functions beyond the cell, serving as signaling molecules in the blood. Researchers have thoroughly investigated the roles of these circulating miRNAs in various diseases. The capacity to detect and quantify ci-miRNAs in bodily fluids suggests their potential as biomarkers for monitoring several health conditions, including cancer, heart disease, brain disorders, and metabolic disorders, where fluctuations in miRNA levels may correlate with different physiological and pathological states. Current methods enable researchers to identify and measure miRNAs in these fluids, facilitating the exploration of their roles in health maintenance and disease resistance. Although research on ci-miRNAs is ongoing, recent studies focus on uncovering their significance, assessing their viability as biomarkers, and clarifying their functions. However, our understanding of how various types, intensities, and durations of exercise influence the levels of these miRNAs in the bloodstream is still limited. This section seeks to provide an overview of the changes in ci-miRNAs in response to exercise.
Collapse
Affiliation(s)
- Xiu Li
- Shanghai Minyuan College, Shanghai 201210, China.
| | - Jamal Hallajzadeh
- Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
2
|
Senyigit A, Durmus S, Oruc A, Gelisgen R, Uzun H, Tabak O. Dysfunction of PTEN-Associated MicroRNA Regulation: Exploring Potential Pathological Links in Type 1 Diabetes Mellitus. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1744. [PMID: 39596932 PMCID: PMC11595949 DOI: 10.3390/medicina60111744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 11/29/2024]
Abstract
Background and Objectives: Type 1 Diabetes Mellitus (T1DM) is an autoimmune disease with T cell-mediated pathogenesis of pancreatic β-cell destruction, leading to insulin deficiency. MicroRNAs such as miR-223 and miR-106b, along with PTEN, have been reported to participate in the pathophysiology of diabetes and its complications. The current study has explored the expression of miR-223, miR-106b, and PTEN and their association with various clinical and biochemical parameters in subjects diagnosed with T1DM. Materials and Methods: Sixty T1DM patients (two groups as uncomplicated/ with microalbuminuria) and fifty healthy volunteers, age- and sex-matched, were enrolled in this study. The fasting venous blood samples were collected, and PTEN and miRNAs (miR-223 and miR-106b) levels were measured by ELISA and real-time PCR, respectively. Results: The PTEN levels of patients with microalbuminuria were significantly lower than those of patients without microalbuminuria, while those of miR-223 and miR-106b were significantly increased in the T1DM group compared with the healthy control group (p < 0.001). ROC analysis indicated that PTEN, miR-223, and miR-106b could be potential biomarkers for diagnosing T1DM with high specificity but with variable sensitivities. Also, PTEN and miR-223 were negatively correlated with r =-0.398 and p < 0.0001, indicating that they were interrelated in their role within the T1DM pathophysiology. Conclusions: In the current study, it has been shown that the circulating levels of PTEN, miR-223, and miR-106b are significantly changed in T1DM patients and may back their potential to be used as non-invasive biomarkers for the diagnosis and monitoring of T1DM. Low PTEN protein expression was related to high miR-223 expression, indicating involvement of these miRNA in the regulation of PTEN. Further studies should be performed to clarify the exact mechanisms and possible clinical applications of these molecules.
Collapse
Affiliation(s)
- Abdulhalim Senyigit
- Department of Internal Medicine, Faculty of Medicine, Istanbul Atlas University, Istanbul 34403, Türkiye;
| | - Sinem Durmus
- Department of Medical Biochemistry, Faculty of Medicine, İzmir Kâtip Celebi University, Izmir 35620, Türkiye;
| | - Aykut Oruc
- Department of Physiology, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34320, Türkiye;
| | - Remise Gelisgen
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul 34320, Türkiye;
| | - Hafize Uzun
- Department of Medical Biochemistry, Faculty of Medicine, Istanbul Atlas University, Istanbul 34403, Türkiye
| | - Omur Tabak
- Department of Internal Medicine, Kanuni Sultan Suleyman Training and Research Hospital, Health Sciences University, Istanbul 34668, Türkiye
| |
Collapse
|
3
|
Odimegwu CL, Uwaezuoke SN, Chikani UN, Mbanefo NR, Adiele KD, Nwolisa CE, Eneh CI, Ndiokwelu CO, Okpala SC, Ogbuka FN, Odo KE, Ohuche IO, Obiora-Izuka CE. Targeting the Epigenetic Marks in Type 2 Diabetes Mellitus: Will Epigenetic Therapy Be a Valuable Adjunct to Pharmacotherapy? Diabetes Metab Syndr Obes 2024; 17:3557-3576. [PMID: 39323929 PMCID: PMC11423826 DOI: 10.2147/dmso.s479077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 08/03/2024] [Indexed: 09/27/2024] Open
Abstract
Although genetic, environmental, and lifestyle factors largely contribute to type 2 diabetes mellitus (T2DM) risk, the role of epigenetics in its pathogenesis is now well established. The epigenetic mechanisms in T2DM mainly consist of DNA methylation, histone modifications and regulation by noncoding RNAs (ncRNAs). For instance, DNA methylation at CpG islands in the promoter regions of specific genes encoding insulin signaling and glucose metabolism suppresses these genes. Modulating the enzyme mediators of these epigenetic marks aims to restore standard gene expression patterns and improve glycemic control. In targeting these epigenetic marks, using epigenetic drugs such as DNA methyltransferase (DNAMT), histone deacetylase (HDAC) and histone acetyltransferase (HAT) inhibitors has led to variable success in humans and experimental murine models. Specifically, the United States' Food and Drug Administration (US FDA) has approved DNAMT inhibitors like 5-azacytidine and 5-aza-2'-deoxycytidine for use in diabetic retinopathy: a T2DM microvascular complication. These DNAMT inhibitors block the genes for methylation of mitochondrial superoxide dismutase 2 (SOD2) and matrix metallopeptidase 9 (MMP-9): the epigenetic marks in diabetic retinopathy. Traditional pharmacotherapy with metformin also have epigenetic effects in T2DM and positively alter disease outcomes when combined with epigenetic drugs like DNAMT and HDAC inhibitors, raising the prospect of using epigenetic therapy as a valuable adjunct to pharmacotherapy. However, introducing small interfering RNAs (siRNAs) in cells to silence specific target genes remains in the exploratory phase. Future research should focus on regulating gene expression in T2DM using long noncoding RNA (lncRNA) molecules, another type of ncRNA. This review discusses the epigenetics of T2DM and that of its macro- and microvascular complications, and the potential benefits of combining epigenetic therapy with pharmacotherapy for optimal results.
Collapse
Affiliation(s)
- Chioma Laura Odimegwu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Samuel Nkachukwu Uwaezuoke
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ugo N Chikani
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ngozi Rita Mbanefo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Ken Daberechi Adiele
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | - Chizoma Ihuarula Eneh
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Chibuzo Obiora Ndiokwelu
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Somkenechi C Okpala
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | - Francis N Ogbuka
- Department of Pediatrics, Enugu State University Teaching Hospital (ESUTH), Enugu, Nigeria
| | - Kenneth E Odo
- Department of Pediatrics, the University of Nigeria Teaching Hospital (UNTH), Ituku-Ozalla Enugu, Nigeria
| | | | | |
Collapse
|
4
|
Voisin S, Seale K, Jacques M, Landen S, Harvey NR, Haupt LM, Griffiths LR, Ashton KJ, Coffey VG, Thompson JM, Doering TM, Lindholm ME, Walsh C, Davison G, Irwin R, McBride C, Hansson O, Asplund O, Heikkinen AE, Piirilä P, Pietiläinen KH, Ollikainen M, Blocquiaux S, Thomis M, Coletta DK, Sharples AP, Eynon N. Exercise is associated with younger methylome and transcriptome profiles in human skeletal muscle. Aging Cell 2024; 23:e13859. [PMID: 37128843 PMCID: PMC10776126 DOI: 10.1111/acel.13859] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/03/2023] Open
Abstract
Exercise training prevents age-related decline in muscle function. Targeting epigenetic aging is a promising actionable mechanism and late-life exercise mitigates epigenetic aging in rodent muscle. Whether exercise training can decelerate, or reverse epigenetic aging in humans is unknown. Here, we performed a powerful meta-analysis of the methylome and transcriptome of an unprecedented number of human skeletal muscle samples (n = 3176). We show that: (1) individuals with higher baseline aerobic fitness have younger epigenetic and transcriptomic profiles, (2) exercise training leads to significant shifts of epigenetic and transcriptomic patterns toward a younger profile, and (3) muscle disuse "ages" the transcriptome. Higher fitness levels were associated with attenuated differential methylation and transcription during aging. Furthermore, both epigenetic and transcriptomic profiles shifted toward a younger state after exercise training interventions, while the transcriptome shifted toward an older state after forced muscle disuse. We demonstrate that exercise training targets many of the age-related transcripts and DNA methylation loci to maintain younger methylome and transcriptome profiles, specifically in genes related to muscle structure, metabolism, and mitochondrial function. Our comprehensive analysis will inform future studies aiming to identify the best combination of therapeutics and exercise regimes to optimize longevity.
Collapse
Affiliation(s)
- Sarah Voisin
- Institute for Health and Sport (iHeS)Victoria UniversityFootscrayVictoriaAustralia
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Kirsten Seale
- Institute for Health and Sport (iHeS)Victoria UniversityFootscrayVictoriaAustralia
| | - Macsue Jacques
- Institute for Health and Sport (iHeS)Victoria UniversityFootscrayVictoriaAustralia
| | - Shanie Landen
- Institute for Health and Sport (iHeS)Victoria UniversityFootscrayVictoriaAustralia
| | - Nicholas R. Harvey
- Faculty of Health Sciences and MedicineBond UniversityGold CoastQueenslandAustralia
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical SciencesQueensland University of TechnologyBrisbaneQueenslandAustralia
| | - Larisa M. Haupt
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical SciencesQueensland University of TechnologyBrisbaneQueenslandAustralia
- ARC Training Centre for Cell and Tissue Engineering TechnologiesQueensland University of Technology (QUT)BrisbaneQueenslandAustralia
- Max Planck Queensland Centre for the Materials Sciences of Extracellular MatricesBrisbaneQueenslandAustralia
| | - Lyn R. Griffiths
- Genomics Research Centre, Centre for Genomics and Personalised Health, School of Biomedical SciencesQueensland University of TechnologyBrisbaneQueenslandAustralia
| | - Kevin J. Ashton
- Faculty of Health Sciences and MedicineBond UniversityGold CoastQueenslandAustralia
| | - Vernon G. Coffey
- Faculty of Health Sciences and MedicineBond UniversityGold CoastQueenslandAustralia
| | | | - Thomas M. Doering
- School of Health, Medical and Applied SciencesCentral Queensland UniversityRockhamptonQueenslandAustralia
| | - Malene E. Lindholm
- Department of Medicine, School of MedicineStanford UniversityStanfordCaliforniaUSA
| | - Colum Walsh
- Genomic Medicine Research Group, School of Biomedical SciencesUlster UniversityColeraineUK
| | - Gareth Davison
- Sport and Exercise Sciences Research InstituteUlster UniversityBelfastUK
| | - Rachelle Irwin
- Genomic Medicine Research Group, School of Biomedical SciencesUlster UniversityColeraineUK
| | - Catherine McBride
- Sport and Exercise Sciences Research InstituteUlster UniversityBelfastUK
| | - Ola Hansson
- Department of Clinical Sciences, Genomics, Diabetes and Endocrinology Unit, Lund University Diabetes CenterLund UniversityLundSweden
- Institute for Molecular Medicine Finland (FIMM)Helsinki UniversityHelsinkiFinland
| | - Olof Asplund
- Department of Clinical Sciences, Genomics, Diabetes and Endocrinology Unit, Lund University Diabetes CenterLund UniversityLundSweden
| | - Aino E. Heikkinen
- Institute for Molecular Medicine Finland (FIMM)Helsinki UniversityHelsinkiFinland
| | - Päivi Piirilä
- Unit of Clinical PhysiologyHelsinki University Hospital and University of HelsinkiHelsinkiFinland
| | - Kirsi H. Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of MedicineUniversity of HelsinkiHelsinkiFinland
- HealthyWeightHub, Endocrinology, Abdominal CenterHelsinki University Hospital and University of HelsinkiHelsinkiFinland
| | - Miina Ollikainen
- Institute for Molecular Medicine Finland (FIMM)Helsinki UniversityHelsinkiFinland
- Minerva Foundation Institute for Medical ResearchHelsinkiFinland
| | - Sara Blocquiaux
- Department of Movement Sciences, Physical Activity, Sports and Health Research GroupKU LeuvenLeuvenBelgium
| | - Martine Thomis
- Department of Movement Sciences, Physical Activity, Sports and Health Research GroupKU LeuvenLeuvenBelgium
| | - Dawn K. Coletta
- Department of Medicine, Division of EndocrinologyUniversity of ArizonaTucsonArizonaUSA
- UA Center for Disparities in Diabetes Obesity and MetabolismUniversity of ArizonaTucsonArizonaUSA
- Department of PhysiologyUniversity of ArizonaTucsonArizonaUSA
| | - Adam P. Sharples
- Institute of Physical PerformanceNorwegian School of Sport SciencesOsloNorway
| | - Nir Eynon
- Institute for Health and Sport (iHeS)Victoria UniversityFootscrayVictoriaAustralia
- Australian Regenerative Medicine InstituteMonash UniversityClaytonVictoriaAustralia
| |
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
|
Masson SWC, Madsen S, Cooke KC, Potter M, Vegas AD, Carroll L, Thillainadesan S, Cutler HB, Walder KR, Cooney GJ, Morahan G, Stöckli J, James DE. Leveraging genetic diversity to identify small molecules that reverse mouse skeletal muscle insulin resistance. eLife 2023; 12:RP86961. [PMID: 37494090 PMCID: PMC10371229 DOI: 10.7554/elife.86961] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023] Open
Abstract
Systems genetics has begun to tackle the complexity of insulin resistance by capitalising on computational advances to study high-diversity populations. 'Diversity Outbred in Australia (DOz)' is a population of genetically unique mice with profound metabolic heterogeneity. We leveraged this variance to explore skeletal muscle's contribution to whole-body insulin action through metabolic phenotyping and skeletal muscle proteomics of 215 DOz mice. Linear modelling identified 553 proteins that associated with whole-body insulin sensitivity (Matsuda Index) including regulators of endocytosis and muscle proteostasis. To enrich for causality, we refined this network by focusing on negatively associated, genetically regulated proteins, resulting in a 76-protein fingerprint of insulin resistance. We sought to perturb this network and restore insulin action with small molecules by integrating the Broad Institute Connectivity Map platform and in vitro assays of insulin action using the Prestwick chemical library. These complementary approaches identified the antibiotic thiostrepton as an insulin resistance reversal agent. Subsequent validation in ex vivo insulin-resistant mouse muscle and palmitate-induced insulin-resistant myotubes demonstrated potent insulin action restoration, potentially via upregulation of glycolysis. This work demonstrates the value of a drug-centric framework to validate systems-level analysis by identifying potential therapeutics for insulin resistance.
Collapse
Affiliation(s)
- Stewart WC Masson
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Søren Madsen
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Kristen C Cooke
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Meg Potter
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Alexis Diaz Vegas
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Luke Carroll
- Australian Proteome Analysis Facility, Macquarie UniversityMacquarie ParkAustralia
| | - Senthil Thillainadesan
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Harry B Cutler
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Ken R Walder
- School of Medicine, Deakin UniversityGeelongAustralia
| | - Gregory J Cooney
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical ResearchMurdochAustralia
| | - Jacqueline Stöckli
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
| | - David E James
- Charles Perkins Centre, School of Life and Environmental Sciences, University of SydneyCamperdownAustralia
- School of Medical Sciences University of SydneySydneyAustralia
| |
Collapse
|
7
|
Oh J, Riek AE, Bauerle KT, Dusso A, McNerney KP, Barve RA, Darwech I, Sprague JE, Moynihan C, Zhang RM, Kutz G, Wang T, Xing X, Li D, Mrad M, Wigge NM, Castelblanco E, Collin A, Bambouskova M, Head RD, Sands MS, Bernal-Mizrachi C. Embryonic vitamin D deficiency programs hematopoietic stem cells to induce type 2 diabetes. Nat Commun 2023; 14:3278. [PMID: 37311757 PMCID: PMC10264405 DOI: 10.1038/s41467-023-38849-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/19/2023] [Indexed: 06/15/2023] Open
Abstract
Environmental factors may alter the fetal genome to cause metabolic diseases. It is unknown whether embryonic immune cell programming impacts the risk of type 2 diabetes in later life. We demonstrate that transplantation of fetal hematopoietic stem cells (HSCs) made vitamin D deficient in utero induce diabetes in vitamin D-sufficient mice. Vitamin D deficiency epigenetically suppresses Jarid2 expression and activates the Mef2/PGC1a pathway in HSCs, which persists in recipient bone marrow, resulting in adipose macrophage infiltration. These macrophages secrete miR106-5p, which promotes adipose insulin resistance by repressing PIK3 catalytic and regulatory subunits and down-regulating AKT signaling. Vitamin D-deficient monocytes from human cord blood have comparable Jarid2/Mef2/PGC1a expression changes and secrete miR-106b-5p, causing adipocyte insulin resistance. These findings suggest that vitamin D deficiency during development has epigenetic consequences impacting the systemic metabolic milieu.
Collapse
Affiliation(s)
- Jisu Oh
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Amy E Riek
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin T Bauerle
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, VA Medical Center, St. Louis, MO, USA
| | - Adriana Dusso
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyle P McNerney
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ruteja A Barve
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Isra Darwech
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Clare Moynihan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rong M Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Greta Kutz
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Marguerite Mrad
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas M Wigge
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Alejandro Collin
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Monika Bambouskova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Bernal-Mizrachi
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Medicine, VA Medical Center, St. Louis, MO, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
8
|
Stokes T, Cen HH, Kapranov P, Gallagher IJ, Pitsillides AA, Volmar C, Kraus WE, Johnson JD, Phillips SM, Wahlestedt C, Timmons JA. Transcriptomics for Clinical and Experimental Biology Research: Hang on a Seq. ADVANCED GENETICS (HOBOKEN, N.J.) 2023; 4:2200024. [PMID: 37288167 PMCID: PMC10242409 DOI: 10.1002/ggn2.202200024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Indexed: 06/09/2023]
Abstract
Sequencing the human genome empowers translational medicine, facilitating transcriptome-wide molecular diagnosis, pathway biology, and drug repositioning. Initially, microarrays are used to study the bulk transcriptome; but now short-read RNA sequencing (RNA-seq) predominates. Positioned as a superior technology, that makes the discovery of novel transcripts routine, most RNA-seq analyses are in fact modeled on the known transcriptome. Limitations of the RNA-seq methodology have emerged, while the design of, and the analysis strategies applied to, arrays have matured. An equitable comparison between these technologies is provided, highlighting advantages that modern arrays hold over RNA-seq. Array protocols more accurately quantify constitutively expressed protein coding genes across tissue replicates, and are more reliable for studying lower expressed genes. Arrays reveal long noncoding RNAs (lncRNA) are neither sparsely nor lower expressed than protein coding genes. Heterogeneous coverage of constitutively expressed genes observed with RNA-seq, undermines the validity and reproducibility of pathway analyses. The factors driving these observations, many of which are relevant to long-read or single-cell sequencing are discussed. As proposed herein, a reappreciation of bulk transcriptomic methods is required, including wider use of the modern high-density array data-to urgently revise existing anatomical RNA reference atlases and assist with more accurate study of lncRNAs.
Collapse
Affiliation(s)
- Tanner Stokes
- Faculty of ScienceMcMaster UniversityHamiltonL8S 4L8Canada
| | - Haoning Howard Cen
- Life Sciences InstituteUniversity of British ColumbiaVancouverV6T 1Z3Canada
| | | | - Iain J Gallagher
- School of Applied SciencesEdinburgh Napier UniversityEdinburghEH11 4BNUK
| | | | | | | | - James D. Johnson
- Life Sciences InstituteUniversity of British ColumbiaVancouverV6T 1Z3Canada
| | | | | | - James A. Timmons
- Miller School of MedicineUniversity of MiamiMiamiFL33136USA
- William Harvey Research InstituteQueen Mary University LondonLondonEC1M 6BQUK
- Augur Precision Medicine LTDStirlingFK9 5NFUK
| |
Collapse
|
9
|
Chang YC, Chan MH, Yang YF, Li CH, Hsiao M. Glucose transporter 4: Insulin response mastermind, glycolysis catalyst and treatment direction for cancer progression. Cancer Lett 2023; 563:216179. [PMID: 37061122 DOI: 10.1016/j.canlet.2023.216179] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/27/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
The glucose transporter family (GLUT) consists of fourteen members. It is responsible for glucose homeostasis and glucose transport from the extracellular space to the cell cytoplasm to further cascade catalysis. GLUT proteins are encoded by the solute carrier family 2 (SLC2) genes and are members of the major facilitator superfamily of membrane transporters. Moreover, different GLUTs also have their transporter kinetics and distribution, so each GLUT member has its uniqueness and importance to play essential roles in human physiology. Evidence from many studies in the field of diabetes showed that GLUT4 travels between the plasma membrane and intracellular vesicles (GLUT4-storage vesicles, GSVs) and that the PI3K/Akt pathway regulates this activity in an insulin-dependent manner or by the AMPK pathway in response to muscle contraction. Moreover, some published results also pointed out that GLUT4 mediates insulin-dependent glucose uptake. Thus, dysfunction of GLUT4 can induce insulin resistance, metabolic reprogramming in diverse chronic diseases, inflammation, and cancer. In addition to the relationship between GLUT4 and insulin response, recent studies also referred to the potential upstream transcription factors that can bind to the promoter region of GLUT4 to regulating downstream signals. Combined all of the evidence, we conclude that GLUT4 has shown valuable unknown functions and is of clinical significance in cancers, which deserves our in-depth discussion and design compounds by structure basis to achieve therapeutic effects. Thus, we intend to write up a most updated review manuscript to include the most recent and critical research findings elucidating how and why GLUT4 plays an essential role in carcinogenesis, which may have broad interests and impacts on this field.
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Hsien Chan
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Fang Yang
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung, Taiwan
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan; Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| |
Collapse
|
10
|
Beilerli A, Kudriashov V, Sufianov A, Kostin A, Begliarzade S, Ilyasova T, Liang Y, Mukhamedzyanov A, Beylerli O. Regulation and mechanism of action of miRNAs on insulin resistance in skeletal muscles. Noncoding RNA Res 2023; 8:218-223. [PMID: 36860209 PMCID: PMC9969252 DOI: 10.1016/j.ncrna.2023.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/14/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
The term "insulin resistance" is commonly understood as a decrease in the response of insulin-sensitive tissues to insulin at its sufficient concentration, leading to chronic compensatory hyperinsulinemia. Type 2 diabetes mellitus is based on mechanisms consisting in the development of resistance to insulin in target cells (hepatocytes, adipocytes, skeletal muscle cells), resulting in the termination of an adequate response of these tissues to interaction with insulin. Since in healthy people 75-80% of glucose is utilized by skeletal muscle, it is more likely that the main cause of insulin resistance is impaired insulin-stimulated glucose utilization by skeletal muscle. With insulin resistance, skeletal muscles do not respond to insulin at its normal concentration, thereby determining an increase in glucose levels and a compensatory increase in insulin production in response to this. Despite many years of studying diabetes mellitus (DM) and insulin resistance, the molecular genetic basis for the development of these pathological conditions is still the subject of numerous studies. Recent studies point to the involvement of microRNAs (miRNAs) as dynamic modifiers in the pathogenesis of various diseases. MiRNAs are a separate class of RNA molecules that play a key role in the post-transcriptional regulation of gene expression. Recent studies have shown that miRNAs dysregulation in DM is closely related to miRNAs regulatory abilities in skeletal muscle insulin resistance. This gave grounds to consider an increase or decrease in the expression of individual microRNAs in muscle tissue and consider them as new biomarkers for diagnosing and monitoring insulin resistance and promising directions for targeted therapy. This review presents the results of scientific studies examining the role of miRNAs in skeletal muscle insulin resistance.
Collapse
Affiliation(s)
- Aferin Beilerli
- Department of Obstetrics and Gynecology, Tyumen State Medical University, 54 Odesskaya Street, 625023, Tyumen, Russia
| | | | - Albert Sufianov
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Department of Neurosurgery, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Andrey Kostin
- Research and Educational Resource Center for Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis Innovative Technologies, Peoples' Friendship University of Russia, Moscow, Russia
| | - Sema Begliarzade
- Republican Clinical Perinatal Center, Ufa, Republic of Bashkortostan, 450106, Russia
| | - Tatiana Ilyasova
- Department of Internal Diseases, Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Yanchao Liang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, China
| | | | - Ozal Beylerli
- Educational and Scientific Institute of Neurosurgery, Рeoples’ Friendship University of Russia (RUDN University), Moscow, Russia
- Corresponding author. Рeoples’ Friendship University of Russia (RUDN University), 6 Miklukho-Maklaya Street, Moscow, 117198, Russian Federation.
| |
Collapse
|
11
|
Taylor HJ, Hung YH, Narisu N, Erdos MR, Kanke M, Yan T, Grenko CM, Swift AJ, Bonnycastle LL, Sethupathy P, Collins FS, Taylor DL. Human pancreatic islet microRNAs implicated in diabetes and related traits by large-scale genetic analysis. Proc Natl Acad Sci U S A 2023; 120:e2206797120. [PMID: 36757889 PMCID: PMC9963967 DOI: 10.1073/pnas.2206797120] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 01/11/2023] [Indexed: 02/10/2023] Open
Abstract
Genetic studies have identified ≥240 loci associated with the risk of type 2 diabetes (T2D), yet most of these loci lie in non-coding regions, masking the underlying molecular mechanisms. Recent studies investigating mRNA expression in human pancreatic islets have yielded important insights into the molecular drivers of normal islet function and T2D pathophysiology. However, similar studies investigating microRNA (miRNA) expression remain limited. Here, we present data from 63 individuals, the largest sequencing-based analysis of miRNA expression in human islets to date. We characterized the genetic regulation of miRNA expression by decomposing the expression of highly heritable miRNAs into cis- and trans-acting genetic components and mapping cis-acting loci associated with miRNA expression [miRNA-expression quantitative trait loci (eQTLs)]. We found i) 84 heritable miRNAs, primarily regulated by trans-acting genetic effects, and ii) 5 miRNA-eQTLs. We also used several different strategies to identify T2D-associated miRNAs. First, we colocalized miRNA-eQTLs with genetic loci associated with T2D and multiple glycemic traits, identifying one miRNA, miR-1908, that shares genetic signals for blood glucose and glycated hemoglobin (HbA1c). Next, we intersected miRNA seed regions and predicted target sites with credible set SNPs associated with T2D and glycemic traits and found 32 miRNAs that may have altered binding and function due to disrupted seed regions. Finally, we performed differential expression analysis and identified 14 miRNAs associated with T2D status-including miR-187-3p, miR-21-5p, miR-668, and miR-199b-5p-and 4 miRNAs associated with a polygenic score for HbA1c levels-miR-216a, miR-25, miR-30a-3p, and miR-30a-5p.
Collapse
Affiliation(s)
- Henry J. Taylor
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, CambridgeCB2 0BB, UK
- Heart and Lung Research Institute, University of Cambridge, CambridgeCB2 0BB, UK
| | - Yu-Han Hung
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY14853
| | - Narisu Narisu
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Michael R. Erdos
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Matthew Kanke
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY14853
| | - Tingfen Yan
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Caleb M. Grenko
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Amy J. Swift
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Lori L. Bonnycastle
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - Praveen Sethupathy
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY14853
| | - Francis S. Collins
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| | - D. Leland Taylor
- Center for Precision Health Research, National Human Genome Research Institute, NIH, Bethesda, MD20892
| |
Collapse
|
12
|
Yang X, Wang K, Lang J, Guo D, Gao H, Qiu Y, Jin X, Zhang M, Shi J, Ma Q, Ma Q, Wen Z. Up-regulation of miR-133a-3p promotes ovary insulin resistance on granulosa cells of obese PCOS patients via inhibiting PI3K/AKT signaling. BMC Womens Health 2022; 22:412. [PMID: 36209087 PMCID: PMC9548189 DOI: 10.1186/s12905-022-01994-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 09/26/2022] [Indexed: 11/05/2022] Open
Abstract
Background MicroRNAs are a type of non-coding single-stranded RNA, which is involved in the regulation of ovary insulin resistance (IR). This study aims to explore the underlying mechanisms of miR-133a-3p regulating ovary IR in obese polycystic ovary syndrome (PCOS).
Methods Granulosa cells (GCs) were extracted from follicular fluids of PCOS patients (obese PCOS group and non-obese PCOS group) and healthy women (control group). The expression of miR-133a-3p in GCs was detected by qRT-PCR. The targets and pathways of miR-133a-3p were predicted by bioinformatics analyses. The protein levels of PI3K, p-AKT, GLUT4, p-GSK-3β, and p-FOXO1 were measured by Western blotting. Results MiR-133a-3p was highly expressed in GCs from PCOS patients, especially in obese PCOS patients. The protein levels of PI3K and p-AKT was downregulated in GCs from PCOS patients. There were 11 target genes of miR-133a-3p enriching in PI3K/AKT signaling pathway. miR-133a-3p mimic downregulated the expression of PI3K, p-AKT, and GLUT4, and upregulated the protein levels of p-GSK-3β and p-FOXO1. miR-133a-3p inhibitor presented the opposite effect of miR-133a-3p mimic. Conclusion MiR-133a-3p promotes ovary IR on GCs of obese PCOS patients via inhibiting PI3K/AKT signaling pathway. This study lays a foundation for further research on the mechanism of ovary IR in obese PCOS patients.
Collapse
Affiliation(s)
- Xiaoman Yang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Kehua Wang
- grid.479672.9Integrative Medicine Center for Reproductive and Heredity, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42 Wenhuaxi Road, Jinan, China
| | - Jiajia Lang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Danyang Guo
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Haixia Gao
- grid.479672.9Integrative Medicine Center for Reproductive and Heredity, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42 Wenhuaxi Road, Jinan, China
| | - Yue Qiu
- grid.479672.9Integrative Medicine Center for Reproductive and Heredity, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42 Wenhuaxi Road, Jinan, China
| | - Xiaohan Jin
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Mingyue Zhang
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiaxiu Shi
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - QianQian Ma
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qian Ma
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zixi Wen
- grid.464402.00000 0000 9459 9325Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Sheng CY, Son YH, Jang J, Park SJ. In vitro skeletal muscle models for type 2 diabetes. BIOPHYSICS REVIEWS 2022; 3:031306. [PMID: 36124295 PMCID: PMC9478902 DOI: 10.1063/5.0096420] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Type 2 diabetes mellitus, a metabolic disorder characterized by abnormally elevated blood sugar, poses a growing social, economic, and medical burden worldwide. The skeletal muscle is the largest metabolic organ responsible for glucose homeostasis in the body, and its inability to properly uptake sugar often precedes type 2 diabetes. Although exercise is known to have preventative and therapeutic effects on type 2 diabetes, the underlying mechanism of these beneficial effects is largely unknown. Animal studies have been conducted to better understand the pathophysiology of type 2 diabetes and the positive effects of exercise on type 2 diabetes. However, the complexity of in vivo systems and the inability of animal models to fully capture human type 2 diabetes genetics and pathophysiology are two major limitations in these animal studies. Fortunately, in vitro models capable of recapitulating human genetics and physiology provide promising avenues to overcome these obstacles. This review summarizes current in vitro type 2 diabetes models with focuses on the skeletal muscle, interorgan crosstalk, and exercise. We discuss diabetes, its pathophysiology, common in vitro type 2 diabetes skeletal muscle models, interorgan crosstalk type 2 diabetes models, exercise benefits on type 2 diabetes, and in vitro type 2 diabetes models with exercise.
Collapse
Affiliation(s)
- Christina Y. Sheng
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | - Young Hoon Son
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | - Sung-Jin Park
- Biohybrid Systems Group, Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia 30322, USA
| |
Collapse
|
14
|
Wu SE, Hsu JC, Chang YL, Chuang HC, Chiu YL, Chen WL. Benzo[a]pyrene exposure in muscle triggers sarcopenia through aryl hydrocarbon receptor-mediated reactive oxygen species production. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 239:113599. [PMID: 35567930 DOI: 10.1016/j.ecoenv.2022.113599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 04/29/2022] [Accepted: 04/30/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Benzo[a]pyrene (BaP), a toxic carcinogen, is associated with various adverse effects but is rarely discussed in muscle-related disorders. This study investigated in vitro and in vivo effects triggered by BaP exposure in muscles and hypothesized that exposure might induce conditions similar to sarcopenia due to the shared mechanism of oxidative stress. In vitro experiments used C2C12 mouse myoblasts to examine effects induced by BaP exposure in control (untreated) and BaP-treated (10 µM/ml) muscle cells. An established TNF-α-treated sarcopenia model was utilized to verify our results. In vivo experiments compared immunohistochemical staining of sarcopenia-related markers in rats exposed to clean air and polluted air. RESULTS In C2C12 cells, after 2-72 h of BaP exposure, elevated mRNA and protein expressions were observed in aryl hydrocarbon receptor (AhR) and cytochrome P450 1A1, subsequently in ROS (NOX2 and NOX4) production, inflammatory cytokines (IL-6, TNF-α, and NF-kB), and proteins mediating apoptotic cell death (caspase-3 and PARP). Two myokines also altered mRNA and protein expressions akin to changes in sarcopenia, namely decreased irisin levels and increased myostatin levels. In addition, N-acetylcysteine, a well-known antioxidant, led to decrease in oxidative markers induced by BaP. The validation by TNF-α-treated sarcopenia model revealed comparable biological responses in either TNF-α or BaP treated C2C12 cells. In vivo experiments with rats exposed to air pollution showed increased expression of BaP, AhR, 8-hydroxydeoxyguanosine, and myostatin and decreased irisin expression in immunohistochemical staining. CONCLUSIONS Our results suggest that BaP exerts deleterious effects on the muscle, leading to conditions indicative of sarcopenia. Antioxidant supplementation may be a treatment option for BaP-induced sarcopenia, but further validation studies are needed.
Collapse
Affiliation(s)
- Shou-En Wu
- Department of Dermatology, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei City, Taiwan (R.O.C); Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei City, Taiwan (R.O.C); Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan (R.O.C)
| | - Ju-Chun Hsu
- Department of Biochemistry, National Defense Medical Center, Taiwan (R.O.C)
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taiwan (R.O.C)
| | - Hsiao-Chi Chuang
- School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan (R.O.C); Division of Pulmonary Medicine, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan (R.O.C); Cell Physiology and Molecular Image Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan (R.O.C)
| | - Yi-Lin Chiu
- Department of Biochemistry, National Defense Medical Center, Taiwan (R.O.C)
| | - Wei-Liang Chen
- Division of Family Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, School of Medicine, National Defense Medical Center, Taipei City, Taiwan (R.O.C); Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital, Taipei, Taiwan (R.O.C); Department of Biochemistry, National Defense Medical Center, Taiwan (R.O.C).
| |
Collapse
|
15
|
Overview of Transcriptomic Research on Type 2 Diabetes: Challenges and Perspectives. Genes (Basel) 2022; 13:genes13071176. [PMID: 35885959 PMCID: PMC9319211 DOI: 10.3390/genes13071176] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/04/2023] Open
Abstract
Type 2 diabetes (T2D) is a common chronic disease whose etiology is known to have a strong genetic component. Standard genetic approaches, although allowing for the detection of a number of gene variants associated with the disease as well as differentially expressed genes, cannot fully explain the hereditary factor in T2D. The explosive growth in the genomic sequencing technologies over the last decades provided an exceptional impetus for transcriptomic studies and new approaches to gene expression measurement, such as RNA-sequencing (RNA-seq) and single-cell technologies. The transcriptomic analysis has the potential to find new biomarkers to identify risk groups for developing T2D and its microvascular and macrovascular complications, which will significantly affect the strategies for early diagnosis, treatment, and preventing the development of complications. In this article, we focused on transcriptomic studies conducted using expression arrays, RNA-seq, and single-cell sequencing to highlight recent findings related to T2D and challenges associated with transcriptome experiments.
Collapse
|
16
|
Zhang S, Liu X, Wang J, Yuan F, Liu Y. Targeting ferroptosis with miR-144-3p to attenuate pancreatic β cells dysfunction via regulating USP22/SIRT1 in type 2 diabetes. Diabetol Metab Syndr 2022; 14:89. [PMID: 35761309 PMCID: PMC9235078 DOI: 10.1186/s13098-022-00852-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Recently, ferroptosis has been implicated in the pathologic process of several diseases including type 2 diabetes mellitus (T2DM). However, molecular mechanisms underlying ferroptosis in T2DM remain obscure. METHODS Twenty four mice were included in this study. T2DM model mice were established by a high-fat diet combined with streptozotocin injection. INS-1 cells were stimulated with high glucose (HG). Cell viability was detected by CCK-8 kit. The levels of GSH, MDA, iron, and lipid ROS, and SOD activity, were detected by the corresponding kits. The interaction between miR-144-3p and USP22 was validated by dual-luciferase reporter assay. The relationship between USP22 and its substrate was verified using Co-IP and ubiquitination assays. The mRNA and protein expressions were examined by RT-qPCR and western blot, respectively. The functions of β cells in vitro and in vivo were evaluated glucose-stimulated insulin secretion test and HOMA-β, respectively. RESULTS Ferroptosis occurred in the pancreas of T2DM mice and HG-induced INS-1 cells. Silencing miR-144-3p blocked the effect of HG on the cell viability and accumulation of lipid peroxides, thereby improving the insulin secretion in INS-1 cells. Mechanistically, USP22 is a direct target of miR-144-3p, which could stabilize SIRT1 expression, thereby suppressing ferroptosis. Overexpressing USP22 attenuated deleterious roles of HG in INS-1 cells; but its roles were reversed by up-regulating miR-144-3p. In vivo study demonstrated that miR-144-3p antagomir exerted an anti-hyperglycemic effect and regulated the ferroptosis-related proteins in the pancreas. CONCLUSION The up-regulation of miR-144-3p suppressed USP22/SIRT1 to induce ferroptosis, which causes pancreatic β cells dysfunction, thereby promoting T2DM development.
Collapse
Affiliation(s)
- Shanshan Zhang
- School of Medicine of Pingdingshan University, Middle Section of Chongwen Road, Xincheng District, Pingdingshan, 467000 Henan China
- Shool of Nursing, Doctor of Philosophy in Nursing, Philippine Women’s University, Manila, Philippines
| | - Xiao Liu
- School of Medicine of Pingdingshan University, Middle Section of Chongwen Road, Xincheng District, Pingdingshan, 467000 Henan China
- Shool of Nursing, Doctor of Philosophy in Nursing, Philippine Women’s University, Manila, Philippines
| | - Jihong Wang
- School of Medicine of Pingdingshan University, Middle Section of Chongwen Road, Xincheng District, Pingdingshan, 467000 Henan China
| | - Fengjuan Yuan
- School of Medicine of Pingdingshan University, Middle Section of Chongwen Road, Xincheng District, Pingdingshan, 467000 Henan China
- Shool of Nursing, Doctor of Philosophy in Nursing, Philippine Women’s University, Manila, Philippines
| | - Yali Liu
- School of Medicine of Pingdingshan University, Middle Section of Chongwen Road, Xincheng District, Pingdingshan, 467000 Henan China
- Shool of Nursing, Doctor of Philosophy in Nursing, Philippine Women’s University, Manila, Philippines
| |
Collapse
|
17
|
Sabaratnam R, Skov V, Paulsen SK, Juhl S, Kruse R, Hansen T, Halkier C, Kristensen JM, Vind BF, Richelsen B, Knudsen S, Dahlgaard J, Beck-Nielsen H, Kruse TA, Højlund K. A Signature of Exaggerated Adipose Tissue Dysfunction in Type 2 Diabetes Is Linked to Low Plasma Adiponectin and Increased Transcriptional Activation of Proteasomal Degradation in Muscle. Cells 2022; 11:cells11132005. [PMID: 35805088 PMCID: PMC9265693 DOI: 10.3390/cells11132005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 06/12/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
Insulin resistance in skeletal muscle in type 2 diabetes (T2D) is characterized by more pronounced metabolic and molecular defects than in obesity per se. There is increasing evidence that adipose tissue dysfunction contributes to obesity-induced insulin resistance in skeletal muscle. Here, we used an unbiased approach to examine if adipose tissue dysfunction is exaggerated in T2D and linked to diabetes-related mechanisms of insulin resistance in skeletal muscle. Transcriptional profiling and biological pathways analysis were performed in subcutaneous adipose tissue (SAT) and skeletal muscle biopsies from 17 patients with T2D and 19 glucose-tolerant, age and weight-matched obese controls. Findings were validated by qRT-PCR and western blotting of selected genes and proteins. Patients with T2D were more insulin resistant and had lower plasma adiponectin than obese controls. Transcriptional profiling showed downregulation of genes involved in mitochondrial oxidative phosphorylation and the tricarboxylic-acid cycle and increased expression of extracellular matrix (ECM) genes in SAT in T2D, whereas genes involved in proteasomal degradation were upregulated in the skeletal muscle in T2D. qRT-PCR confirmed most of these findings and showed lower expression of adiponectin in SAT and higher expression of myostatin in muscle in T2D. Interestingly, muscle expression of proteasomal genes correlated positively with SAT expression of ECM genes but inversely with the expression of ADIPOQ in SAT and plasma adiponectin. Protein content of proteasomal subunits and major ubiquitin ligases were unaltered in the skeletal muscle of patients with T2D. A transcriptional signature of exaggerated adipose tissue dysfunction in T2D, compared with obesity alone, is linked to low plasma adiponectin and increased transcriptional activation of proteasomal degradation in skeletal muscle.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 7LE, UK
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, DK-4000 Roskilde, Denmark;
| | - Søren K. Paulsen
- Department of Pathology, Viborg Regional Hospital, DK-8800 Viborg, Denmark;
| | - Stine Juhl
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Rikke Kruse
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Thea Hansen
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Cecilie Halkier
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
| | - Jonas M. Kristensen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Molecular Physiology Section, Department of Nutrition, Exercise and Sports, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Birgitte F. Vind
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
| | - Bjørn Richelsen
- Steno Diabetes Center Aarhus, Aarhus University Hospital, DK-8200 Aarhus N, Denmark;
| | - Steen Knudsen
- Allarity Therapeutics Europe, DK-2970 Hørsholm, Denmark;
| | - Jesper Dahlgaard
- Program for Mind and Body in Mental Health, Research Centre for Health and Welfare Technology, VIA University College, DK-8200 Aarhus, Denmark;
- Department of Clinical Medicine, Aarhus University, DK-8200 Aarhus, Denmark
| | - Henning Beck-Nielsen
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
| | - Torben A. Kruse
- Department of Clinical Genetics, Odense University Hospital, DK-5000 Odense C, Denmark;
| | - Kurt Højlund
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000 Odense C, Denmark; (R.S.); (S.J.); (R.K.); (J.M.K.); (B.F.V.); (H.B.-N.)
- Department of Clinical Research, University of Southern Denmark, DK-5000 Odense C, Denmark; (T.H.); (C.H.)
- Correspondence: ; Tel.: +45-2532-0648
| |
Collapse
|
18
|
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: 0.3] [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
|
19
|
Bourgeois BL, Lin HY, Yeh AY, Levitt DE, Primeaux SD, Ferguson TF, Molina PE, Simon L. Unique circulating microRNA associations with dysglycemia in people living with HIV and alcohol use. Physiol Genomics 2022; 54:36-44. [PMID: 34859690 PMCID: PMC8891241 DOI: 10.1152/physiolgenomics.00085.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
People living with HIV (PLWH) have increased prevalence of comorbid conditions including insulin resistance and at-risk alcohol use. Circulating microRNAs (miRs) may serve as minimally invasive indicators of pathophysiological states. We aimed to identify whether alcohol modulates circulating miR associations with measures of glucose/insulin dynamics in PLWH. PLWH (n = 96; 69.8% males) enrolled in the Alcohol & Metabolic Comorbidities in PLWH: Evidence-Driven Interventions (ALIVE-Ex) study were stratified into negative phosphatidylethanol (PEth < 8 ng/mL, n = 42) and positive PEth (PEth ≥ 8 ng/mL, n = 54) groups. An oral glucose tolerance test (OGTT) was administered, and total RNA was isolated from fasting plasma to determine absolute miR expression. Circulating miRs were selected based on their role in skeletal muscle (miR-133a and miR-206), pancreatic β-cell (miR-375), liver (miR-20a), and adipose tissue (miR-let-7b, miR-146a, and miR-221) function. Correlation and multiple regression analyses between miR expression and adiponectin, 2 h glucose, insulin, and C-peptide values were performed adjusting for body mass index (BMI) category, age, sex, and viral load. miR-133a was negatively associated with adiponectin (P = 0.002) in the negative PEth group, and miR-20a was positively associated with 2 h glucose (P = 0.013) in the positive PEth group. Regression analyses combining miRs demonstrated that miR-133a (P < 0.001) and miR-221 (P = 0.010) together predicted adiponectin in the negative PEth group. miR-20a (P < 0.001) and miR-375 (P = 0.002) together predicted 2 h glucose in the positive PEth group. Our results indicate that associations between miRs and measures of glucose/insulin dynamics differed between PEth groups, suggesting that the pathophysiological mechanisms contributing to altered glucose homeostasis in PLWH are potentially modulated by alcohol use.
Collapse
Affiliation(s)
- Brianna L. Bourgeois
- 1Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana,2Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Hui-Yi Lin
- 2Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana,3School of Public Health, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Alice Y. Yeh
- 1Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Danielle E. Levitt
- 1Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana,2Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Stefany D. Primeaux
- 1Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana,4Joint Diabetes, Endocrinology & Metabolism Program, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana
| | - Tekeda F. Ferguson
- 1Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana,2Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana,5Department of Epidemiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Patricia E. Molina
- 1Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana,2Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| | - Liz Simon
- 1Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, Louisiana,2Comprehensive Alcohol-HIV/AIDS Research Center, Louisiana State University Health Sciences Center, New Orleans, Louisiana
| |
Collapse
|
20
|
Kesharwani D, Kumar A, Poojary M, Scaria V, Datta M. RNA sequencing reveals potential interacting networks between the altered transcriptome and ncRNome in the skeletal muscle of diabetic mice. Biosci Rep 2021; 41:BSR20210495. [PMID: 34190986 PMCID: PMC8276098 DOI: 10.1042/bsr20210495] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 06/21/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
For a global epidemic like Type 2 diabetes mellitus (T2DM), while impaired gene regulation is identified as a primary cause of aberrant cellular physiology; in the past few years, non-coding RNAs (ncRNAs) have emerged as important regulators of cellular metabolism. However, there are no reports of comprehensive in-depth cross-talk between these regulatory elements and the potential consequences in the skeletal muscle during diabetes. Here, using RNA sequencing, we identified 465 mRNAs and 12 long non-coding RNAs (lncRNAs), to be differentially regulated in the skeletal muscle of diabetic mice and pathway enrichment analysis of these altered transcripts revealed pathways of insulin, FOXO and AMP-activated protein kinase (AMPK) signaling to be majorly over-represented. Construction of networks showed that these pathways significantly interact with each other that might underlie aberrant skeletal muscle metabolism during diabetes. Gene-gene interaction network depicted strong interactions among several differentially expressed genes (DEGs) namely, Prkab2, Irs1, Pfkfb3, Socs2 etc. Seven altered lncRNAs depicted multiple interactions with the altered transcripts, suggesting possible regulatory roles of these lncRNAs. Inverse patterns of expression were observed between several of the deregulated microRNAs (miRNAs) and the differentially expressed transcripts in the tissues. Towards validation, overexpression of miR-381-3p and miR-539-5p in skeletal muscle C2C12 cells significantly decreased the transcript levels of their targets, Nfkbia, Pik3r1 and Pi3kr1, Cdkn2d, respectively. Collectively, the findings provide a comprehensive understanding of the interactions and cross-talk between the ncRNome and transcriptome in the skeletal muscle during diabetes and put forth potential therapeutic options for improving insulin sensitivity.
Collapse
Affiliation(s)
- Devesh Kesharwani
- CSIR-Institute of Genomics and Integrative Biology, Functional and Genomics Unit, Mall Road, Delhi, India
- Academy of Scientific and Innovative Research, CSIR-HRDC, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Amit Kumar
- CSIR-Institute of Genomics and Integrative Biology, Functional and Genomics Unit, Mall Road, Delhi, India
- Academy of Scientific and Innovative Research, CSIR-HRDC, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| | - Mukta Poojary
- Academy of Scientific and Innovative Research, CSIR-HRDC, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
- GN Ramachandran Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, Delhi 110025, India
| | - Vinod Scaria
- Academy of Scientific and Innovative Research, CSIR-HRDC, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
- GN Ramachandran Knowledge Centre for Genome Informatics, CSIR-Institute of Genomics and Integrative Biology, Mathura Road, Delhi 110025, India
| | - Malabika Datta
- CSIR-Institute of Genomics and Integrative Biology, Functional and Genomics Unit, Mall Road, Delhi, India
- Academy of Scientific and Innovative Research, CSIR-HRDC, Kamala Nehru Nagar, Ghaziabad 201002, Uttar Pradesh, India
| |
Collapse
|
21
|
Saccon TD, Schneider A, Marinho CG, Nunes ADC, Noureddine S, Dhahbi J, Nunez Lopez YO, LeMunyan G, Salvatori R, Oliveira CRP, Oliveira‐Santos AA, Musi N, Bartke A, Aguiar‐Oliveira MH, Masternak MM. Circulating microRNA profile in humans and mice with congenital GH deficiency. Aging Cell 2021; 20:e13420. [PMID: 34118183 PMCID: PMC8282278 DOI: 10.1111/acel.13420] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/10/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Reduced inflammation, increased insulin sensitivity, and protection against cancer are shared between humans and mice with GH/IGF1 deficiency. Beyond hormone levels, miRNAs are important regulators of metabolic changes associated with healthy aging. We hypothesized that GH deficiency in humans alters the abundance of circulating miRNAs and that a subset of those miRNAs may overlap with those found in GH-deficient mice. In this study, subjects with untreated congenital isolated GH deficiency (IGHD; n = 23) and control subjects matched by age and sex (n = 23) were recruited and serum was collected for miRNA sequencing. Serum miRNAs from young (6 month) and old (22 month) Ames dwarf (df/df) mice with GH deficiency and their WT littermates (n = 5/age/genotype group) were used for comparison. We observed 14 miRNAs regulated with a genotype by age effect and 19 miRNAs regulated with a genotype effect independent of age in serum of IGHD subjects. These regulated miRNAs are known for targeting pathways associated with longevity such as mTOR, insulin signaling, and FoxO. The aging function was overrepresented in IGHD individuals, mediated by hsa-miR-31, hsa-miR-146b, hsa-miR-30e, hsa-miR-100, hsa-miR-181b-2, hsa-miR-195, and hsa-miR-181b-1, which target the FoxO and mTOR pathways. Intriguingly, miR-181b-5p, miR-361-3p, miR-144-3p, and miR-155-5p were commonly regulated in the serum of humans and GH-deficient mice. In vitro assays confirmed target genes for the main up-regulated miRNAs, suggesting miRNAs regulated in IGHD individuals can regulate the expression of age-related genes. These findings indicate that systemic miRNAs regulated in IGHD individuals target pathways involved in aging in both humans and mice.
Collapse
Affiliation(s)
- Tatiana D. Saccon
- Centro de Desenvolvimento Tecnológico Universidade Federal de Pelotas Pelotas Brazil
- Burnett School of Biomedical Sciences College of Medicine University of Central Florida Orlando FL USA
| | - Augusto Schneider
- Faculdade de Nutrição Universidade Federal de Pelotas Pelotas Brazil
| | - Cindi G. Marinho
- Division of Endocrinology Health Sciences Graduate Program Federal University of Sergipe Aracaju Brazil
| | - Allancer D. C. Nunes
- Burnett School of Biomedical Sciences College of Medicine University of Central Florida Orlando FL USA
| | - Sarah Noureddine
- Burnett School of Biomedical Sciences College of Medicine University of Central Florida Orlando FL USA
| | - Joseph Dhahbi
- Department of Medical Education School of Medicine California University of Science & Medicine San Bernardino CA USA
| | - Yury O. Nunez Lopez
- Advent Health Translational Research Institute for Metabolism and Diabetes Orlando FL USA
| | - Gage LeMunyan
- Department of Medical Education School of Medicine California University of Science & Medicine San Bernardino CA USA
| | - Roberto Salvatori
- Division of Endocrinology, Diabetes and Metabolism Department of Medicine The Johns Hopkins University School of Medicine Baltimore MD USA
| | - Carla R. P. Oliveira
- Division of Endocrinology Health Sciences Graduate Program Federal University of Sergipe Aracaju Brazil
| | - Alécia A. Oliveira‐Santos
- Division of Endocrinology Health Sciences Graduate Program Federal University of Sergipe Aracaju Brazil
| | - Nicolas Musi
- Barshop Institute for Longevity and Aging Studies Center for Healthy Aging University of Texas Health Sciences Center at San Antonio and South Texas Veterans Health Care System San Antonio TX USA
- San Antonio Geriatric Research Education and Clinical Center South Texas Veterans Health Care System San Antonio TX USA
| | - Andrzej Bartke
- Department of Internal Medicine Southern Illinois University School of Medicine Springfield IL USA
| | - Manuel H. Aguiar‐Oliveira
- Division of Endocrinology Health Sciences Graduate Program Federal University of Sergipe Aracaju Brazil
| | - Michal M. Masternak
- Burnett School of Biomedical Sciences College of Medicine University of Central Florida Orlando FL USA
- Department of Head and Neck Surgery Poznan University of Medical Sciences Poznan Poland
| |
Collapse
|
22
|
Tonyan ZN, Nasykhova YA, Mikhailova AA, Glotov AS. MicroRNAs as Potential Biomarkers of Type 2 Diabetes Mellitus. RUSS J GENET+ 2021. [DOI: 10.1134/s1022795421060107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
23
|
Dial AG, Monaco CMF, Grafham GK, Romanova N, Simpson JA, Tarnopolsky MA, Perry CGR, Kalaitzoglou E, Hawke TJ. Muscle and serum myostatin expression in type 1 diabetes. Physiol Rep 2021; 8:e14500. [PMID: 32652899 PMCID: PMC7354085 DOI: 10.14814/phy2.14500] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/02/2020] [Accepted: 06/03/2020] [Indexed: 12/17/2022] Open
Abstract
Type 1 diabetes (T1D) has been reported to negatively affect the health of skeletal muscle, though the underlying mechanisms are unknown. Myostatin, a myokine whose increased expression is associated with muscle‐wasting diseases, has not been reported in humans with T1D but has been demonstrated to be elevated in preclinical diabetes models. Thus, the purpose of this study was to determine if there is an elevated expression of myostatin in the serum and skeletal muscle of persons with T1D compared to controls. Secondarily, we aimed to explore relationships between myostatin expression and clinically important metrics (e.g., HbA1c, strength, lean mass) in women and men with (N = 31)/without T1D (N = 24) between 18 and 72 years old. Body composition, baseline strength, blood sample and vastus lateralis muscle biopsy were evaluated. Serum, but not muscle, myostatin expression was significantly elevated in those with T1D versus controls, and to a greater degree in T1D women than T1D men. Serum myostatin levels were not significantly associated with HbA1c nor disease duration. A significant correlation between serum myostatin expression and maximal voluntary contraction (MVC) and body fat mass was demonstrated in control subjects, but these correlations did not reach significance in those with T1D (MVC: R = 0.64 controls vs. R = 0.37 T1D; Body fat: R = −0.52 controls/R = −0.02 T1D). Collectively, serum myostatin was correlated with lean mass (R = 0.45), and while this trend was noted in both groups separately, neither reached statistical significance (R = 0.47 controls/R = 0.33 T1D). Overall, while those with T1D exhibited elevated serum myostatin levels (particularly females) myostatin expression was not correlated with clinically relevant metrics despite some of these relationships existing in controls (e.g., lean/fat mass). Future studies will be needed to fully understand the mechanisms underlying increased myostatin in T1D, with relationships to insulin dosing being particularly important to elucidate.
Collapse
Affiliation(s)
- Athan G Dial
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Cynthia M F Monaco
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Grace K Grafham
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| | - Nadya Romanova
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | | | | | - Evangelia Kalaitzoglou
- Barnstable Brown Diabetes Center and Department of Pediatrics, University of Kentucky, Lexington, KY, USA
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
24
|
Chen YY, Chiu YL, Kao TW, Peng TC, Yang HF, Chen WL. Cross-sectional associations among P3NP, HtrA, Hsp70, Apelin and sarcopenia in Taiwanese population. BMC Geriatr 2021; 21:192. [PMID: 33743591 PMCID: PMC7980650 DOI: 10.1186/s12877-021-02146-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 03/10/2021] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Sarcopenia is a multifactorial pathophysiologic condition of skeletal muscle mass and muscle strength associated with aging. However, biomarkers for predicting the occurrence of sarcopenia are rarely discussed in recent studies. The aim of the study was to elucidate the relationship between sarcopenia and several pertinent biomarkers. METHODS Using the Gene Expression Omnibus (GEO) profiles of the National Center for Biotechnology Information, the associations between mRNA expression of biomarkers and sarcopenia were explored, including high temperature requirement serine protease A1 (HtrA1), procollagen type III N-terminal peptide (P3NP), apelin, and heat shock proteins 70 (Hsp72). We enrolled 408 community-dwelling adults aged 65 years and older with sarcopenia and nonsarcopenia based on the algorithm proposed by the Asian Working Group for Sarcopenia (AWGS). Muscle strength is identified by hand grip strength using an analogue isometric dynamometer. Muscle mass is estimated by skeletal mass index (SMI) using a bioelectrical impedance analysis. Physical performance is measured by gait speed using 6 m walking distance. The associations between these biomarkers and sarcopenia were determined using receiver operating characteristic (ROC) curve analysis and multivariate regression models. RESULTS From the GEO profiles, the sarcopenia gene set variation analysis score was correlated significantly with the mRNA expression of APLNR (p < 0.001) and HSPA2 (p < 0.001). In our study, apelin was significantly associated with decreased hand grip strength with β values of - 0.137 (95%CI: - 0.229, - 0.046) in men. P3NP and HtrA1 were significantly associated with increased SMI with β values of 0.081 (95%CI: 0.010, 0.153) and 0.005 (95%CI: 0.001, 0.009) in men, respectively. Apelin and HtrA1 were inversely associated with the presence of sarcopenia with an OR of 0.543 (95%CI: 0.397-0.743) and 0.003 (95%CI: 0.001-0.890) after full adjustment. The cutoff point of HtrA1 was associated with the presence of sarcopenia with an OR of 0.254 (95%CI: 0.083-0.778) in men. The cutoff point of apelin was negatively associated with the presence of sarcopenia with an OR of 0.254 (95%CI: 0.083-0.778). CONCLUSION Our study highlights that P3NP, HtrA, and apelin are useful for diagnosis of sarcopenia in the clinical setting.
Collapse
Affiliation(s)
- Yuan-Yuei Chen
- Department of Pathology, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Department of Pathology, Tri-Service General Hospital Songshan Branch; and School of Medicine, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China
| | - Yi-Lin Chiu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - Tung-Wei Kao
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China
| | - Tao-Chun Peng
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China
| | - Hui-Fang Yang
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China
| | - Wei-Liang Chen
- Division of Geriatric Medicine, Department of Family and Community Medicine, Tri-Service General Hospital; and School of Medicine, National Defense Medical Center, Number 325, Section 2, Chang-gong Rd, Nei-Hu District, 114, Taipei, Taiwan, Republic of China.
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan, Republic of China.
| |
Collapse
|
25
|
da Paixão AO, Bolin AP, Silvestre JG, Rodrigues AC. Palmitic Acid Impairs Myogenesis and Alters Temporal Expression of miR-133a and miR-206 in C2C12 Myoblasts. Int J Mol Sci 2021; 22:ijms22052748. [PMID: 33803124 PMCID: PMC7963199 DOI: 10.3390/ijms22052748] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/26/2022] Open
Abstract
Palmitic acid (PA), a saturated fatty acid enriched in high-fat diet, has been implicated in the development of sarcopenic obesity. Herein, we chose two non-cytotoxic concentrations to better understand how excess PA could impact myotube formation or diameter without inducing cell death. Forty-eight hours of 100 µM PA induced a reduction of myotube diameter and increased the number of type I fibers, which was associated with increased miR-206 expression. Next, C2C12 myotube growth in the presence of PA was evaluated. Compared to control cells, 150 µM PA reduces myoblast proliferation and the expression of MyoD and miR-206 and miR-133a expression, leading to a reduced number and diameter of myotubes. PA (100 µM), despite not affecting proliferation, impairs myotube formation by reducing the expression of Myf5 and miR-206 and decreasing protein synthesis. Interestingly, 100 and 150 µM PA-treated myotubes had a higher number of type II fibers than control cells. In conclusion, PA affects negatively myotube diameter, fusion, and metabolism, which may be related to myomiRs. By providing new insights into the mechanisms by which PA affects negatively skeletal muscle, our data may help in the discovery of new targets to treat sarcopenic obesity.
Collapse
Affiliation(s)
- Ailma O. da Paixão
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (A.O.d.P.); (A.P.B.)
| | - Anaysa Paola Bolin
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (A.O.d.P.); (A.P.B.)
| | - João G. Silvestre
- Department of Anatomy, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil;
| | - Alice Cristina Rodrigues
- Department of Pharmacology, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo 05508-000, Brazil; (A.O.d.P.); (A.P.B.)
- Correspondence: ; Tel.: +55-11-3091-7406
| |
Collapse
|
26
|
Villota SD, Toledo-Rodriguez M, Leach L. Compromised barrier integrity of human feto-placental vessels from gestational diabetic pregnancies is related to downregulation of occludin expression. Diabetologia 2021; 64:195-210. [PMID: 33001231 PMCID: PMC7716932 DOI: 10.1007/s00125-020-05290-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/18/2020] [Indexed: 12/16/2022]
Abstract
AIMS/HYPOTHESIS Reduced occupancy of junctional occludin is a feature of human placental vessels in the diabetic milieu. However, the functional consequence of this and whether this loss is due to differential expression of occludin splice variants is not known. Our study aimed to investigate the effects of gestational diabetes mellitus (GDM), and its treatment, on endothelial junctional integrity, gene and protein expression of occludin splice variants, and potential regulation of expression by microRNAs (miRNAs). METHODS Term placentas were obtained from normal pregnancies (n = 21), and pregnancies complicated by GDM where glucose levels were controlled by diet (n = 11) or metformin (n = 6). Gene and microRNA (miRNA) expression were determined by quantitative real-time PCR; protein expression by immunoblotting; endothelial junctional occupancy by fluorescence microscopy and systematic sampling; and paracellular leakage by perfusion of placental microvascular beds with 76 Mr dextran. Transfection studies of miRNAs that target OCLN were performed in HUVECs, and the trans-endothelial electrical resistance and tracer permeability of the HUVECs were measured. RESULTS All three predicted OCLN gene splice variants and two occludin protein isoforms were found in human placental samples. In placental samples from diet-controlled GDM (d-GDM) pregnancies we found a lower percentage of conduit vessels showing occludin immunoreactivity (12%, p < 0.01), decreased levels of the fully functional occludin isoform-A protein (29%), and differential gene expression of OCLN variant 2 (33% decrease), variant 3 (3.3-fold increase). These changes were not seen in samples from the group with metformin-controlled GDM. In d-GDM placentas, increased numbers of conduit microvessels demonstrated extravasation of 76 Mr dextran (2.0-fold). In d-GDM expression of one of the five potential miRNAs targeting OCLN, miR-181a-5p, expression was 2.1-fold that in normal pregnancies. Experimental overexpression of miR-181a-5p in HUVECs from normal pregnancies resulted in a highly significant downregulation of OCLN variant 1 (69%) and variant 2 (46%) gene expression, with decreased trans-endothelial resistance (78%) and increase in tracer permeability (1.3-fold). CONCLUSIONS/INTERPRETATION Downregulation of expression of OCLN variant 2 and the fully functional occludin isoform-A protein are a feature of placentas in d-GDM pregnancies. These may be behind the loss of junctional occludin and the increased extravasation of exogenous dextran observed. miR-181a-5p was in part responsible for the downregulation of occludin in placentas from d-GDM pregnancies. Induced overexpression of miR-181a-5p compromised the integrity of the endothelial barrier. Our data suggest that, despite good glucose control, the adoption of lifestyle changes alone during a GDM pregnancy may not be enough to prevent an alteration in the expression of occludin and the subsequent functional consequences in placentas and impaired vascular barrier function in offspring. Graphical abstract.
Collapse
Affiliation(s)
| | | | - Lopa Leach
- School of Life Sciences, University of Nottingham, Nottingham, UK.
| |
Collapse
|
27
|
Simaitis S, Schulte-Körne B, Schiffer T, Bloch W, Predel HG, Brixius K, Brinkmann C. Evidence for Training-Induced Changes in miRNA Levels in the Skeletal Muscle of Patients With Type 2 Diabetes Mellitus. Front Physiol 2020; 11:599651. [PMID: 33343393 PMCID: PMC7744661 DOI: 10.3389/fphys.2020.599651] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/05/2020] [Indexed: 01/13/2023] Open
Abstract
Physical training can improve glycemic control in patients with type 2 diabetes mellitus (T2DM). However, the underlying mechanisms are not entirely clear. An interesting piece of the puzzle could be the regulation of micro-RNAs (miRNAs). They are important modulators of protein expression. Some miRNAs were found to be both linked to poor glycemic control/insulin resistance (with evidence from in vivo and/or in vitro studies) and dysregulated in the skeletal muscle of T2DM patients. This pilot study examines whether a 3-month endurance training program [three times a week, 70-80% peak heart rate (HRpeak)] can down-regulate their levels in T2DM men (n = 7). One skeletal muscle biopsy sample was obtained from each patient at T1 (6 weeks pre-intervention), one at T2 (1 week pre-intervention) and one at T3 (3-4 days post-intervention). miRNA-27a-3p, -29a-3p, -29b-3p, -29c-3p, -106b-5p, -135a-5p, -143-3p, -144-3p, -194-5p, and - 206 levels were determined by RT-qPCR. Friedman ANOVA and post-hoc tests showed that miRNA-29b-3p, -29c-3p and -135a-5p levels were significantly reduced post-training (T3 vs. T2 and/or T1). Glycated hemoglobin (HbA1c) and HOMA insulin resistance index did not change significantly. However, HbA1c was reduced in 6 of 7 patients post-training. Furthermore, Spearman's rank correlation analyses with all values from all time points showed significant negative associations between miRNA-29c-3p, -106b-5p, -144-3p and -194-5p levels and cardiorespiratory fitness (VO2peak). The study results imply that regular exercise and improving one's physical fitness is helpful for the regulation of skeletal muscle miRNAs in T2DM patients. Whether or not changes in the miRNA profile can affect the clinical situation of T2DM patients warrants further research.
Collapse
Affiliation(s)
- Sarah Simaitis
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Benedikt Schulte-Körne
- Department of Preventive and Rehabilitative Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Thorsten Schiffer
- Outpatient Clinic for Sports Traumatology and Public Health Consultation, German Sport University Cologne, Cologne, Germany
| | - Wilhelm Bloch
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Hans-Georg Predel
- Department of Preventive and Rehabilitative Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Klara Brixius
- Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany
| | - Christian Brinkmann
- Department of Preventive and Rehabilitative Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne, Cologne, Germany.,IST University of Applied Sciences, Düsseldorf, Germany
| |
Collapse
|
28
|
García-García FJ, Monistrol-Mula A, Cardellach F, Garrabou G. Nutrition, Bioenergetics, and Metabolic Syndrome. Nutrients 2020; 12:E2785. [PMID: 32933003 PMCID: PMC7551996 DOI: 10.3390/nu12092785] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/05/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
According to the World Health Organization (WHO), the global nutrition report shows that whilst part of the world's population starves, the other part suffers from obesity and associated complications. A balanced diet counterparts these extreme conditions with the proper proportion, composition, quantity, and presence of macronutrients, micronutrients, and bioactive compounds. However, little is known on the way these components exert any influence on our health. These nutrients aiming to feed our bodies, our tissues, and our cells, first need to reach mitochondria, where they are decomposed into CO2 and H2O to obtain energy. Mitochondria are the powerhouse of the cell and mainly responsible for nutrients metabolism, but they are also the main source of oxidative stress and cell death by apoptosis. Unappropriated nutrients may support mitochondrial to become the Trojan horse in the cell. This review aims to provide an approach to the role that some nutrients exert on mitochondria as a major contributor to high prevalent Western conditions including metabolic syndrome (MetS), a constellation of pathologic conditions which promotes type II diabetes and cardiovascular risk. Clinical and experimental data extracted from in vitro animal and cell models further demonstrated in patients, support the idea that a balanced diet, in a healthy lifestyle context, promotes proper bioenergetic and mitochondrial function, becoming the best medicine to prevent the onset and progression of MetS. Any advance in the prevention and management of these prevalent complications help to face these challenging global health problems, by ameliorating the quality of life of patients and reducing the associated sociosanitary burden.
Collapse
Affiliation(s)
- Francesc Josep García-García
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Anna Monistrol-Mula
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Francesc Cardellach
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| | - Glòria Garrabou
- Muscle Research and Mitochondrial Function Laboratory, CELLEX-IDIBAPS, Internal Medicine Department, Faculty of Medicine, University of Barcelona, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (F.J.G.-G.); (A.M.-M.); (F.C.)
- CIBERER—Centre for Biomedical Research Network in Rare Diseases, 28029 Madrid, Spain
| |
Collapse
|
29
|
Non-Coding RNA: Role in Gestational Diabetes Pathophysiology and Complications. Int J Mol Sci 2020; 21:ijms21114020. [PMID: 32512799 PMCID: PMC7312670 DOI: 10.3390/ijms21114020] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 05/30/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
Gestational Diabetes Mellitus (GDM) is defined as glucose intolerance that develops in the second or third trimester of pregnancy. GDM can lead to short-term and long-term complications both in the mother and in the offspring. Diagnosing and treating this condition is therefore of great importance to avoid poor pregnancy outcomes. There is increasing interest in finding new markers with potential diagnostic, prognostic and therapeutic utility in GDM. Non-coding RNAs (ncRNAs), including microRNAs, long non-coding RNAs and circular RNAs, are critically involved in metabolic processes and their dysregulated expression has been reported in several pathological contexts. The aberrant expression of several circulating or placenta-related ncRNAs has been linked to insulin resistance and β-cell dysfunction, the key pathophysiological features of GDM. Furthermore, significant associations between altered ncRNA profiles and GDM-related complications, such as macrosomia or trophoblast dysfunction, have been observed. Remarkably, the deregulation of ncRNAs, which might be linked to a detrimental intrauterine environment, can lead to changes in the expression of target genes in the offspring, possibly contributing to the development of long-term GDM-related complications, such as metabolic and cardiovascular diseases. In this review, all the recent findings on ncRNAs and GDM are summarized, particularly focusing on the molecular aspects and the pathophysiological implications of this complex relationship.
Collapse
|
30
|
Gottmann P, Ouni M, Zellner L, Jähnert M, Rittig K, Walther D, Schürmann A. Polymorphisms in miRNA binding sites involved in metabolic diseases in mice and humans. Sci Rep 2020; 10:7202. [PMID: 32350386 PMCID: PMC7190857 DOI: 10.1038/s41598-020-64326-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 04/02/2020] [Indexed: 02/07/2023] Open
Abstract
Type 2 diabetes and obesity are well-studied metabolic diseases, which are based on genetic and epigenetic alterations in combination with an obesogenic lifestyle. The aim of this study was to test whether SNPs in miRNA-mRNA binding sites that potentially disrupt binding, elevate the expression of miRNA targets, which participate in the development of metabolic diseases. A computational approach was developed that integrates transcriptomics, linkage analysis, miRNA-target prediction data, and sequence information of a mouse model of obesity and diabetes. A statistical analysis demonstrated a significant enrichment of 566 genes for a location in obesity- and diabetes-related QTL. They are expressed at higher levels in metabolically relevant tissues presumably due to altered miRNA-mRNA binding sites. Of these, 51 genes harbor conserved and impaired miRNA-mRNA-interactions in human. Among these, 38 genes have been associated to metabolic diseases according to the phenotypes of corresponding knockout mice or other results described in the literature. The remaining 13 genes (e.g. Jrk, Megf9, Slfn8 and Tmem132e) could be interesting candidates and will be investigated in the future.
Collapse
Affiliation(s)
- Pascal Gottmann
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München, Neuherberg, Germany
| | - Meriem Ouni
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München, Neuherberg, Germany
| | - Lisa Zellner
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München, Neuherberg, Germany
| | - Markus Jähnert
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, 14558, Nuthetal, Germany.,German Center for Diabetes Research (DZD), 85764, München, Neuherberg, Germany
| | - Kilian Rittig
- Clinic for Angiology and Diabetology, 15236, Frankfurt (Oder), Germany.,University of Potsdam, Institute of Nutritional Sciences, Nuthetal, Germany
| | - Dirk Walther
- Max Planck Institute of Molecular Plant Physiology, Am Mühlenberg 1, 14476, Potsdam, Golm, Germany
| | - Annette Schürmann
- German Institute of Human Nutrition Potsdam-Rehbruecke, Department of Experimental Diabetology, 14558, Nuthetal, Germany. .,German Center for Diabetes Research (DZD), 85764, München, Neuherberg, Germany. .,University of Potsdam, Institute of Nutritional Sciences, Nuthetal, Germany. .,Faculty of Health Sciences, joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, the Brandenburg Medical School Theodor Fontane and the University of Potsdam, Potsdam, Germany.
| |
Collapse
|
31
|
Wander PL, Enquobahrie DA, Bammler TK, Srinouanprachanh S, MacDonald J, Kahn SE, Leonetti D, Fujimoto WY, Boyko EJ. Short Report: Circulating microRNAs are associated with incident diabetes over 10 years in Japanese Americans. Sci Rep 2020; 10:6509. [PMID: 32300167 PMCID: PMC7162904 DOI: 10.1038/s41598-020-63606-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/02/2020] [Indexed: 02/07/2023] Open
Abstract
Epigenetic changes precede the development of diabetes by many years, providing clues to its pathogenesis. We explored whether the epigenetic markers, circulating microRNAs (miRNAs), were associated with incident diabetes in Japanese Americans. We conducted a pilot study (n = 10) using plasma from age- and sex-matched participants who did or did not develop diabetes in the Japanese American Community Diabetes Study, an observational study of diabetes risk factors. Extraction and high-throughput sequencing of miRNAs were performed using samples collected at baseline. Regression models were fit comparing circulating miRNAs (N = 1640) among individuals who did or did not develop incident diabetes at 10-year follow-up. Participants averaged 51.7 years of age at baseline; 60% were male. We identified 36 miRNAs present at different (10 higher and 26 lower) levels in individuals who developed diabetes compared to those who did not (log2fold change ≥1.25 and false discovery rate ≤5%). These included miRNAs with functions in skeletal muscle insulin metabolism (miR-106b and miR-20b-5p) and miRNAs with functions in both skeletal muscle insulin metabolism and cell cycle regulation in endocrine pancreas (miR-15a and miR-17). Circulating miRNAs were associated with subsequent development of diabetes among Japanese Americans over 10 years of follow-up. Results are preliminary. Large-scale miRNA sequencing studies could inform our understanding of diabetes pathogenesis and development of therapies, based on gene expression regulation, that target diabetes.
Collapse
Affiliation(s)
- Pandora L Wander
- Department of Medicine,University of Washington, Seattle, WA, United States.
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States.
| | | | - Theo K Bammler
- Department of Environmental and Occupational Health Sciences,University of Washington, Seattle, WA, United States
| | - Sengkeo Srinouanprachanh
- Department of Environmental and Occupational Health Sciences,University of Washington, Seattle, WA, United States
| | - James MacDonald
- Department of Environmental and Occupational Health Sciences,University of Washington, Seattle, WA, United States
| | - Steven E Kahn
- Department of Medicine,University of Washington, Seattle, WA, United States
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| | - Donna Leonetti
- Department of Anthropology, University of Washington, Seattle, WA, United States
| | - Wilfred Y Fujimoto
- Department of Medicine,University of Washington, Seattle, WA, United States
| | - Edward J Boyko
- Department of Medicine,University of Washington, Seattle, WA, United States
- Veterans Affairs Puget Sound Health Care System, Seattle, WA, United States
| |
Collapse
|
32
|
Hess AL, Larsen LH, Udesen PB, Sanz Y, Larsen TM, Dalgaard LT. Levels of Circulating miR-122 are Associated with Weight Loss and Metabolic Syndrome. Obesity (Silver Spring) 2020; 28:493-501. [PMID: 32090516 DOI: 10.1002/oby.22704] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 10/14/2019] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study investigated whether the levels of specific serum microRNAs (miRNAs) were altered following diet-induced weight loss and whether the serum miRNAs differed in the presence of the metabolic syndrome. METHODS The study was a weight loss intervention trial with a prescribed energy deficit of approximately 500 kcal/d. Levels of 22 miRNAs were determined in serum samples from 85 participants with overweight or obesity. miRNAs were analyzed using TaqMan Array miRNA Cards and normalized to the geometric mean of spiked-in ath-miR-159a and U6 small nuclear RNA using the ΔCT method. RESULTS The average weight loss was 5.7 kg (P < 0.001). miR-122-5p (-0.18 ± 0.06 log fold relative to initial, P < 0.01) and miR-193a-5p (-0.12 ± 0.04, P < 0.01) levels decreased in response to weight loss. miR-126a-3p (0.11 ± 0.04, P = 0.01) and miR-222-3p (1.51 ± 0.12, P < 0.001) levels increased. Furthermore, a higher level of miR-122-5p was observed at baseline in participants with the metabolic syndrome compared with participants without (0.28 ± 0.08, P < 0.01). CONCLUSIONS Changes in circulating miR-122-5p, miR-126a-3p, miR-193a-5p, and miR-222-3p in response to diet-induced weight loss are demonstrated. Furthermore, assessment of miR-122-5p could be an indicator of an adverse metabolic health status independent of obesity.
Collapse
Affiliation(s)
- Anne Lundby Hess
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Lesli Hingstrup Larsen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | - Pernille Baekgaard Udesen
- Department of Science and Environment, Roskilde University, Roskilde, Denmark
- Department of Gynaecology and Obstetrics, Fertility Clinic, Zealand University Hospital, Køge, Denmark
| | - Yolanda Sanz
- Microbial Ecology, Nutrition and Health Research Unit, Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
| | - Thomas Meinert Larsen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg, Denmark
| | | |
Collapse
|
33
|
Sadeghzadeh S, Dehghani Ashkezari M, Seifati SM, Vahidi Mehrjardi MY, Dehghan Tezerjani M, Sadeghzadeh S, Ladan SAB. Circulating miR-15a and miR-222 as Potential Biomarkers of Type 2 Diabetes. Diabetes Metab Syndr Obes 2020; 13:3461-3469. [PMID: 33061506 PMCID: PMC7537850 DOI: 10.2147/dmso.s263883] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/04/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In recent years, considerable attention has been paid to the role of microRNAs (miRs) as biomarkers in type 2 diabetes (T2D). The aim of the study was to evaluate the expression levels of miR-15a and miR-222 in diabetic, pre-diabetic, and healthy individuals. MATERIALS AND METHODS Ninety individuals, who were referred to the Yazd diabetic center, were enrolled in this study and then classified into three groups as healthy, pre-T2D, and diabetic based on the clinical manifestations. Real-time PCR was performed to explore miRs expression in the plasma samples of the studied population. The correlation between the biochemical characteristic and the expression of these miRs as well as specificity and sensitivity of different clinical markers in healthy and pre-diabetic groups was evaluated. RESULTS miR-222 expression was significantly upregulated in the pre-T2D cases compared to the control subjects (P<0.001), while no significant difference was found between the pre-T2D and T2D groups (P<0.05). The expression of miR-15a was statistically downregulated in the pre-T2D and T2D subjects (P<0.05). The receiver operating characteristic (ROC) curve analysis of miR-15a expression with a cutoff point of 1.12 resulted in the area under the curve (AUC) of 85% (95% CI 0.865-0.912; P<0.001) with 84% and 85% sensitivity and specificity, respectively. Similarly, for miR-222, the cutoff point of 4.03 and AUC of 86% (95% CI 0.875-0.943; P<0.001) discriminated against the pre-T2D and control subjects via the sensitivity and specificity of 86% and 87%, respectively. Moreover, miR-15a values showed a negative correlation with FG (R=-0.32, P=0.005); however, miR-222 values were positively correlated with FG (R=0.25, P=0.03) in the pre-T2D group. Furthermore, miR-222 values were correlated with OGTT in the pre-T2D group (R=0.27, P=0.001). In addition, LDL-C had a negative correlation with miR-222 values in the pre-T2D group (R=-0.23, P=0.02). CONCLUSION This study indicated that the plasma expression levels of miR-222 and miR-15a can be considered as non-invasive, fast tools to separate the pre-T2D individuals from their healthy counterparts. Accordingly, this information could be used to predict the development of the disease as well as a direction for optimal therapy, thus refining outcomes in patients with diabetes.
Collapse
Affiliation(s)
- Salman Sadeghzadeh
- Medical Biotechnology Research Center, Ashkezar Branch, Islamic Azad University, Ashkezar, Yazd, Iran
| | - Mahmood Dehghani Ashkezari
- Medical Biotechnology Research Center, Ashkezar Branch, Islamic Azad University, Ashkezar, Yazd, Iran
- Correspondence: Mahmood Dehghani Ashkezari Medical Biotechnology Research Center, Ashkezar Branch, Islamic Azad University, Ashkezar, Yazd, Iran Email
| | - Seyed Morteza Seifati
- Medical Biotechnology Research Center, Ashkezar Branch, Islamic Azad University, Ashkezar, Yazd, Iran
| | - Mohammad Yahya Vahidi Mehrjardi
- Diabetes Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Genetics, Medical School, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Dehghan Tezerjani
- Abortion Research Center, Yazd Institute of Reproductive Sciences, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sara Sadeghzadeh
- Department of Physiology, Faculty of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Amir Behtash Ladan
- Medical Biotechnology Research Center, Ashkezar Branch, Islamic Azad University, Ashkezar, Yazd, Iran
| |
Collapse
|
34
|
Singh V, Kumar R, Ojha K, Kushwaha P. Muscle biopsies differ in relation to expression of fiber-type specific genes. JOURNAL OF DIABETOLOGY 2020. [DOI: 10.4103/jod.jod_6_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
35
|
Rosado JA, Diez-Bello R, Salido GM, Jardin I. Fine-tuning of microRNAs in Type 2 Diabetes Mellitus. Curr Med Chem 2019; 26:4102-4118. [PMID: 29210640 DOI: 10.2174/0929867325666171205163944] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 11/23/2017] [Accepted: 11/23/2017] [Indexed: 12/13/2022]
Abstract
Type 2 diabetes mellitus is a metabolic disease widely spread across industrialized countries. Sedentary lifestyle and unhealthy alimentary habits lead to obesity, boosting both glucose and fatty acid in the bloodstream and eventually, insulin resistance, pancreas inflammation and faulty insulin production or secretion, all of them very well-defined hallmarks of type 2 diabetes mellitus. miRNAs are small sequences of non-coding RNA that may regulate several processes within the cells, fine-tuning protein expression, with an unexpected and subtle precision and in time-frames ranging from minutes to days. Since the discovery of miRNA and their possible implication in pathologies, several groups aimed to find a relationship between type 2 diabetes mellitus and miRNAs. Here we discuss the pattern of expression of different miRNAs in cultured cells, animal models and diabetic patients. We summarize the role of the most important miRNAs involved in pancreas growth and development, insulin secretion and liver, skeletal muscle or adipocyte insulin resistance in the context of type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Juan A Rosado
- Institute of Molecular Pathology Biomarkers & Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| | - Raquel Diez-Bello
- Institute of Molecular Pathology Biomarkers & Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| | - Ginés M Salido
- Institute of Molecular Pathology Biomarkers & Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| | - Isaac Jardin
- Institute of Molecular Pathology Biomarkers & Department of Physiology (Cell Physiology Research Group), University of Extremadura, 10003-Caceres, Spain
| |
Collapse
|
36
|
Kokkinopoulou I, Maratou E, Mitrou P, Boutati E, Sideris DC, Fragoulis EG, Christodoulou MI. Decreased expression of microRNAs targeting type-2 diabetes susceptibility genes in peripheral blood of patients and predisposed individuals. Endocrine 2019; 66:226-239. [PMID: 31559537 DOI: 10.1007/s12020-019-02062-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 08/20/2019] [Indexed: 12/12/2022]
Abstract
AIM Certain microRNA molecules (miRNAs) that target genes involved in beta-cell growth and insulin resistance are found deregulated in patients with type-2 diabetes mellitus (T2D) and correlate with its complications. However, the expression profile of miRNAs that regulate genes bearing T2D-related single-nucleotide polymorphisms has been hardly studied. We recently reported that the mRNA patterns of specific T2D-susceptibility genes are impaired in patients, and associate with disease parameters and risk factors. The aim of this study was to explore the levels of miRNAs that target those genes, in peripheral blood of patients versus controls. METHODS A panel of 14 miRNAs validated to target the CDKN2A, CDK5, IGF2BP2, KCNQ1, and TSPAN8 genes, was developed upon combined search throughout the DIANNA TarBase v7.0, miRTarBase, miRSearch v3.0-Exiqon, miRGator v3.0, and miRTarget Link Human algorithms. Specifically developed poly(A)polyadenylation(PAP)-reverse transcription(RT)-qPCR protocols were applied in peripheral blood RNA samples from patients and controls. Possible correlations with the disease, clinicopathological parameters and/or risk factors were evaluated. RESULTS T2D patients expressed decreased levels of let-7b-5p, miR-1-3p, miR-24-3p, miR-34a-5p, miR-98-5p, and miR-133a-3p, compared with controls. Moreover, these levels correlated with certain disease features including insulin and % HbA1c levels in patients, as well as BMI, triglycerides' levels and family history in controls. CONCLUSIONS A T2D-specific expression profile of miRNAs that target disease-susceptibility genes is for the first time described. Future studies are needed to elucidate the associated transcription-regulatory mechanisms, perchance involved in T2D pathogenesis, and to evaluate the potential of these molecules as possible biomarkers for this disorder.
Collapse
Affiliation(s)
- Ioanna Kokkinopoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
| | - Eirini Maratou
- Second Department of Internal Medicine and Research Institute, School of Medicine, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | | | - Eleni Boutati
- Second Department of Internal Medicine and Research Institute, School of Medicine, National and Kapodistrian University of Athens, "Attikon" University Hospital, Athens, Greece
| | - Diamantis C Sideris
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Emmanuel G Fragoulis
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Ioanna Christodoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece.
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK.
| |
Collapse
|
37
|
Abstract
BACKGROUND Obesity and type 2 diabetes (T2D) are major public health issues worldwide, and put a significant burden on the healthcare system. Genetic variants, along with traditional risk factors such as diet and physical activity, could account for up to approximately a quarter of disease risk. Epigenetic factors have demonstrated potential in accounting for additional phenotypic variation, along with providing insights into the causal relationship linking genetic variants to phenotypes. SCOPE OF REVIEW In this review article, we discuss the epidemiological and functional insights into epigenetic disturbances in obesity and diabetes, along with future research directions and approaches, with a focus on DNA methylation. MAJOR CONCLUSIONS Epigenetic mechanisms have been shown to contribute to obesity and T2D disease development, as well as potential differences in disease risks between ethnic populations. Technology to investigate epigenetic profiles in diseased individuals and tissues has advanced significantly in the last years, and suggests potential in application of epigenetic factors in clinical monitoring and as therapeutic options.
Collapse
Affiliation(s)
- Marie Loh
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore; Translational Laboratory in Genetic Medicine, Agency for Science, Technology and Research, Singapore (A*STAR) 138648, Singapore; Yong Loo Lin School of Medicine, National University of Singapore 117596, Singapore; Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK
| | - Li Zhou
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore
| | - Hong Kiat Ng
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore
| | - John Campbell Chambers
- Lee Kong Chian School of Medicine, Nanyang Technological University 308232, Singapore; Department of Epidemiology and Biostatistics, Imperial College London, London W2 1PG, UK; Department of Cardiology, Ealing Hospital, London North West Healthcare NHS Trust, Southall UB1 3HW, UK; Imperial College Healthcare NHS Trust, London W12 0HS, UK.
| |
Collapse
|
38
|
Li Y, Li C, Yang M, Shi L, Tao W, Shen K, Li X, Wang X, Yang Y, Yao Y. Association of single nucleotide polymorphisms of miRNAs involved in the GLUT4 pathway in T2DM in a Chinese population. Mol Genet Genomic Med 2019; 7:e907. [PMID: 31389668 PMCID: PMC6732275 DOI: 10.1002/mgg3.907] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Background The insulin/insulin receptor substrate (IRS)/phosphatidylinositol 3‐kinase (PI3K)/protein kinase B (Akt)/GLUT4 pathway plays a crucial role in insulin resistance and is closely associated with T2DM. Accumulating evidence indicates that miRNAs (such as miR‐135a, let‐7d, miR‐107, miR‐96, miR‐29a, miR‐23a, miR‐126, miR‐133a, and miR‐106b) influence the GLUT4 pathway. Methods A total of 784 subjects with T2DM and 846 nondiabetic subjects were enrolled and 12 single nucleotide polymorphisms (SNPs) in miRNAs (rs10459194 in miR‐135a‐2, rs10993081 and rs7045890 in let‐7d, rs2296616 in miR‐107, rs2402959 and rs6965643 in miR‐96, rs24168 in miR‐29a, rs3745453 in miR‐23a, rs4636297 in miR‐126, rs8089787 and rs9948906 in miR‐133a‐1 and rs999885 in miR‐106b) involved in the GLUT4 pathway were genotyped using the MassArray method in a Chinese population. Results Our data showed that the A allele of rs2402959 in miR‐96 may increase the risk of developing T2DM (p = .002, OR = 1.266; 95% CI: 1.089–1.471). The genotypes of rs3745453 in miR‐23a showed the difference between T2DM and control groups (p < .001). Moreover, for rs2402959, compared with the A/A genotype, the (G/A–G/G) genotype shows a protective effect in T2DM (p = .001, OR = 0.71; 95% CI: 0.58–0.87). For rs3745453, compared with the (A/A–A/G) genotype, the G/G genotype increases the risk of T2DM (p < .001, OR = 1.95; 95% CI: 1.38–2.77). In addition, we also found that rs4636297G/G genotype was associated with lower TC in T2DM group. Conclusion Our results revealed that genetic variations in the miRNAs involved in the GLUT4 pathway were associated with T2DM susceptibility in a Chinese population, and these results emphasize the need to study the functional effects of these variations in the miRNAs involved in the GLUT4 pathway on the risk of developing T2DM.
Collapse
Affiliation(s)
- Yiping Li
- Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province & The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Chuanyin Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan, China
| | - Man Yang
- Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province & The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Li Shi
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan, China
| | - Wenyu Tao
- Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province & The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Keyu Shen
- Faculty of Medicine, Dentistry and Healthy Science, The University of Melbourne, Melbourne, Vic., Australia
| | - Xianli Li
- Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province & The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Xiaoling Wang
- Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province & The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ying Yang
- Department of Endocrinology and Metabolism, The Second People's Hospital of Yunnan Province & The Fourth Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Yufeng Yao
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming, Yunnan, China
| |
Collapse
|
39
|
Paseban M, Butler AE, Sahebkar A. Mechanisms of statin‐induced new‐onset diabetes. J Cell Physiol 2019; 234:12551-12561. [DOI: 10.1002/jcp.28123] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 12/20/2018] [Indexed: 08/30/2023]
Abstract
AbstractStatins, with their lipid‐lowering properties, are a first‐line therapy for the prevention of cardiovascular diseases. Recent evidence, however, suggests that statins can increase the risk of new‐onset diabetes (NOD). The molecular mechanisms of statin‐induced NOD are not precisely known, although some pathophysiologic mechanisms have been suggested. Specific to the beta cell, these mechanisms include alterations in insulin secretion, changes in ion channels, modulation of signaling pathways, and inflammation/oxidative stress. Outwith the beta cell, other suggested mechanisms involve adipocytes, including alterations in adipocyte differentiation and modulation of leptin and adiponectin, and genetic and epigenetic mechanisms, including alterations in microRNA. The evidence supporting these and other mechanisms will be discussed. Greater understanding of the underlying mechanisms linking the onset of diabetes to statin therapy is essential and clinically relevant, as it may enable novel preventative or therapeutic approaches to be instituted and guide the production of a new generation of statins lacking this side effect.
Collapse
Affiliation(s)
- Maryam Paseban
- Department of Physiology Faculty of Medicine, Mashhad University of Medical Sciences Mashhad Iran
| | | | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences Mashhad Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences Mashhad Iran
- School of Pharmacy, Mashhad University of Medical Sciences Mashhad Iran
| |
Collapse
|
40
|
Extracellular Vesicle Encapsulated MicroRNAs in Patients with Type 2 Diabetes Are Affected by Metformin Treatment. J Clin Med 2019; 8:jcm8050617. [PMID: 31067715 PMCID: PMC6571700 DOI: 10.3390/jcm8050617] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 01/05/2023] Open
Abstract
Recently, microRNAs (miRNAs) in circulating extracellular vesicles (EVs), have emerged as a source of potential biomarkers for various pathophysiological conditions, including metabolic disorders such as diabetes. Type 2 diabetes mellitus (T2DM), is the most prevalent form of diabetes in the USA, with 30 million diagnosed patients. Identifying miRNA biomarkers that can be used to assess response to glucose lowering treatments would be useful. Using patient plasma samples from a subset of the Danish Metagenomics of the Human Intestinal Tract (MetaHIT) cohort, we characterized miRNAs from whole plasma, plasma-derived EVs, and EV-depleted plasma by small RNA-sequencing to identify T2DM associated miRNAs. We identified several miRNAs that exhibited concentration changes between controls and non-metformin treated T2DM patients and we validated a subset of these by quantitative reverse transcription-polymerase chain reaction (qRT-PCR). The results showed that the concentrations of many T2DM-affected miRNAs in EV (but not in whole or EV-depleted plasma) decreased to levels close to those of healthy controls following metformin treatment. Among other potential uses of these differentially expressed miRNAs, some might be useful in assessing the response to metformin in T2DM patients.
Collapse
|
41
|
Garneau L, Aguer C. Role of myokines in the development of skeletal muscle insulin resistance and related metabolic defects in type 2 diabetes. DIABETES & METABOLISM 2019; 45:505-516. [PMID: 30844447 DOI: 10.1016/j.diabet.2019.02.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/04/2019] [Accepted: 02/25/2019] [Indexed: 12/20/2022]
Abstract
Due to its mass, skeletal muscle is the major site of glucose uptake and an important tissue in the development of type 2 diabetes (T2D). Muscles of patients with T2D are affected with insulin resistance and mitochondrial dysfunction, which result in impaired glucose and fatty acid metabolism. A well-established method of managing the muscle metabolic defects occurring in T2D is physical exercise. During exercise, muscles contract and secrete factors called myokines which can act in an autocrine/paracrine fashion to improve muscle energy metabolism. In patients with T2D, plasma levels as well as muscle levels (mRNA and protein) of some myokines are upregulated, while others are downregulated. The signalling pathways of certain myokines are also altered in skeletal muscle of patients with T2D. Taken together, these findings suggest that myokine secretion is an important factor contributing to the development of muscle metabolic defects during T2D. It is also of interest considering that lack of physical activity is closely linked to the occurrence of this disease. The causal relationships between sedentary behavior, factors secreted by skeletal muscle at rest and during contraction and the development of T2D remain to be elucidated. Many myokines shown to influence muscle energy metabolism still have not been characterized in the context of T2D in skeletal muscle specifically. The purpose of this review is to highlight what is known and what remains to be determined regarding myokine secretion in patients with T2D to uncover potential therapeutic targets for the management of this disease.
Collapse
Affiliation(s)
- L Garneau
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Institut du Savoir Montfort - recherche, Ottawa, ON, K1K 0T2, Canada
| | - C Aguer
- University of Ottawa, Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, Ottawa, ON, K1H 8M5, Canada; Institut du Savoir Montfort - recherche, Ottawa, ON, K1K 0T2, Canada.
| |
Collapse
|
42
|
Houshmand-Oeregaard A, Schrölkamp M, Kelstrup L, Hansen NS, Hjort L, Thuesen ACB, Broholm C, Mathiesen ER, Clausen TD, Vaag A, Damm P. Increased expression of microRNA-15a and microRNA-15b in skeletal muscle from adult offspring of women with diabetes in pregnancy. Hum Mol Genet 2019. [PMID: 29528396 DOI: 10.1093/hmg/ddy085] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Offspring of women with diabetes in pregnancy exhibit skeletal muscle insulin resistance and are at increased risk of developing type 2 diabetes, potentially mediated by epigenetic mechanisms or changes in the expression of small non-coding microRNAs. Members of the miR-15 family can alter the expression or function of important proteins in the insulin signalling pathway, affecting insulin sensitivity and secretion. We hypothesized that exposure to maternal diabetes may cause altered expression of these microRNAs in offspring skeletal muscle, representing a potential underlying mechanism by which exposure to maternal diabetes leads to increased risk of cardiometabolic disease in offspring. We measured microRNA expression in skeletal muscle biopsies of 26- to 35-year-old offspring of women with either gestational diabetes (O-GDM, n = 82) or type 1 diabetes (O-T1DM, n = 67) in pregnancy, compared with a control group of offspring from the background population (O-BP, n = 57) from an observational follow-up study. Expression of both miR-15a and miR-15b was increased in skeletal muscle obtained from O-GDM (both P < 0.001) and O-T1DM (P = 0.024, P = 0.005, respectively) compared with O-BP. Maternal 2 h post OGTT glucose levels were positively associated with miR-15a expression (P = 0.041) in O-GDM after adjustment for confounders and mediators. In all groups collectively, miRNA expression was significantly positively associated with fasting plasma glucose, 2 h plasma glucose and HbA1c. We conclude that fetal exposure to maternal diabetes is associated with increased skeletal muscle expression of miR-15a and miR-15b and that this may contribute to development of metabolic disease in these subjects.
Collapse
Affiliation(s)
- Azadeh Houshmand-Oeregaard
- Department of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, 2100 Copenhagen, Denmark.,Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maren Schrölkamp
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Louise Kelstrup
- Department of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Ninna S Hansen
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Line Hjort
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Danish Diabetes Academy, 5000 Odense, Denmark
| | - Anne Cathrine B Thuesen
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Christa Broholm
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Elisabeth R Mathiesen
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Department of Endocrinology, Center for Pregnant Women with Diabetes, Rigshospitalet, 2100 Copenhagen, Denmark
| | - Tine D Clausen
- Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Department of Gynecology and Obstetrics, Nordsjaellands Hospital, University of Copenhagen, 3400 Hilleroed, Denmark
| | - Allan Vaag
- Department of Endocrinology, Diabetes and Metabolism, Rigshospitalet, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,Cardiovascular and Metabolic Disease (CVMD) Translational Medicine Unit, Early Clinical Development, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Peter Damm
- Department of Obstetrics, Center for Pregnant Women with Diabetes, Rigshospitalet, 2100 Copenhagen, Denmark.,Institute of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
43
|
Campbell-Tofte J, Vrahatis A, Josefsen K, Mehlsen J, Winther K. Investigating the aetiology of adverse events following HPV vaccination with systems vaccinology. Cell Mol Life Sci 2019; 76:67-87. [PMID: 30324425 PMCID: PMC11105185 DOI: 10.1007/s00018-018-2925-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Revised: 09/10/2018] [Accepted: 09/20/2018] [Indexed: 12/18/2022]
Abstract
In contrast to the insidious and poorly immunogenic human papillomavirus (HPV) infections, vaccination with the HPV virus-like particles (vlps) is non-infectious and stimulates a strong neutralizing-antibody response that protects HPV-naïve vaccinees from viral infection and associated cancers. However, controversy about alleged adverse events following immunization (AEFI) with the vlps have led to extensive reductions in vaccine acceptance, with countries like Japan dropping it altogether. The AEFIs are grouped into chronic fatigue syndrome/myalgic encephalomyelitis (CFS/ME). In this review, we present a hypothesis that the AEFIs might arise from malfunctions within the immune system when confronted with the unusual antigen. In addition, we outline how the pathophysiology of the AEFIs can be cost-effectively investigated with the holistic principles of systems vaccinology in a two-step process. First, comprehensive immunological profiles of HPV vaccinees exhibiting the AEFIs are generated by integrating the data derived from serological profiling for prominent HPV antibodies and serum cytokines, with data from serum metabolomics, peripheral white blood cells transcriptomics and gut microbiome profiling. Next, the immunological profiles are compared with corresponding profiles generated for matched (a) HPV vaccinees without AEFIs; (b) non-HPV-vaccinated individuals with CFS/ME-like symptoms; and (c) non-HPV-vaccinated individuals without CFS/ME. In these comparisons, any causal links between HPV vaccine and the AEFIs, as well as the underlying molecular basis for the links will be revealed. Such a study should provide an objective basis for evaluating HPV vaccine safety and for identifying biomarkers for individuals at risk of developing AEFI with HPV vaccination.
Collapse
Affiliation(s)
| | | | - Knud Josefsen
- Bartholin Institute, Rigshospitalet, Blegdamsvej 9, 2100, Copenhagen Ø, Denmark
| | - Jesper Mehlsen
- Coordinating Research Centre, Bispebjerg and Frederiksberg Hospital, Nordre Fasanvej 57, 2000, Frederiksberg, Denmark
| | - Kaj Winther
- Department of Nutrition, Exercise and Sports, University of Copenhagen, Nørre Allé 51, DK-2200, Copenhagen N, Denmark
| |
Collapse
|
44
|
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: 4.7] [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
|
45
|
Håkansson KEJ, Sollie O, Simons KH, Quax PHA, Jensen J, Nossent AY. Circulating Small Non-coding RNAs as Biomarkers for Recovery After Exhaustive or Repetitive Exercise. Front Physiol 2018; 9:1136. [PMID: 30246800 PMCID: PMC6113669 DOI: 10.3389/fphys.2018.01136] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/30/2018] [Indexed: 01/30/2023] Open
Abstract
Circulating microRNAs have proven to be reliable biomarkers, due to their high stability, both in vivo in the circulation, and ex vivo during sample preparation and storage. Small nucleolar RNAs (snoRNAs) are a different type of small non-coding RNAs that can also be reliably measured in plasma, but have only been studied sporadically. In this study, we aimed to identify RNA-biomarkers that can distinguish between different exercise regimes and that entail clues about muscle repair and recovery after prolonged exhaustive endurance exercise. We compared plasma microRNA profiles between two cohorts of elite cyclists, subjected to two different types of exercise regimes, as well as a cohort of patients with peripheral artery disease (PAD) that were scheduled to undergo lower limb amputation, due to critical limb ischemia. In elite athletes, muscle tissue recovers quickly even after exhaustive exercise, whereas in PAD patients, recovery is completely impaired. Furthermore, we measured levels of a specific group of snoRNAs in the plasma of both elite cyclists and PAD patients. Using a multiplex qPCR screening, we detected a total of 179 microRNAs overall, of which, on average, 161 microRNAs were detected per sample. However, only 30 microRNAs were consistently expressed in all samples. Of these, two microRNAs, miR-29a-3p and miR193a-5p, that responded differently two different types of exercise, namely exhaustive exercise and non-exhaustive endurance exercise. Using individual rt/qPCR, we also identified a snoRNA, SNORD114.1, which was significantly upregulated in plasma in response to endurance exercise. Furthermore, two microRNAs, miR-29a-3p and miR-495-3p, were significantly differentially expressed in athletes compared to PAD patients, but only following exercise. We suggest that these two microRNAs could function as markers of impaired muscle repair and recovery. In conclusion, microRNAs miR-29a-3p and miR-193a-5p may help us distinguish between repeated exhaustive and non-exhaustive endurance exercise. MicroRNA miR-29a-3p, as well as miR-495-3p, may further mark impaired muscle recovery in patients with severe critical limb ischemia. Furthermore, we showed for the first time that a circulating snoRNA, SNORD114.1, is regulated in response to exercise and may be used as biomarker.
Collapse
Affiliation(s)
- Kjell E J Håkansson
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Ove Sollie
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - Karin H Simons
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Paul H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands
| | - Jørgen Jensen
- Department of Physical Performance, Norwegian School of Sport Sciences, Oslo, Norway
| | - A Yaël Nossent
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, Netherlands.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, Netherlands.,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria
| |
Collapse
|
46
|
Koltai E, Bori Z, Osvath P, Ihasz F, Peter S, Toth G, Degens H, Rittweger J, Boldogh I, Radak Z. Master athletes have higher miR-7, SIRT3 and SOD2 expression in skeletal muscle than age-matched sedentary controls. Redox Biol 2018; 19:46-51. [PMID: 30107294 PMCID: PMC6092475 DOI: 10.1016/j.redox.2018.07.022] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 07/25/2018] [Accepted: 07/31/2018] [Indexed: 12/13/2022] Open
Abstract
Regular physical exercise has health benefits and can prevent some of the ageing-associated muscle deteriorations. However, the biochemical mechanisms underlying this exercise benefit, especially in human tissues, are not well known. To investigate, we assessed this using miRNA profiling, mRNA and protein levels of anti-oxidant and metabolic proteins in the vastus lateralis muscle of master athletes aged over 65 years and age-matched controls. Master athletes had lower levels of miR-7, while mRNA or protein levels of SIRT3, SIRT1, SOD2, and FOXO1 levels were significantly higher in the vastus lateralis muscle of master athletes compared to muscles of age-matched controls. These results suggest that regular exercise results in better cellular metabolism and antioxidant capacity via maintaining physiological state of mitochondria and efficient ATP production and decreasing ageing-related inflammation as indicated by the lower level of miR-7 in master athletes.
Collapse
Affiliation(s)
- Erika Koltai
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Zoltan Bori
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary
| | - Peter Osvath
- Department of Health Sciences and Sports Medicine, University of Physical Education, Budapest, Hungary
| | - Ferenc Ihasz
- Hungary Institute of Sport Science, Faculty of Education and Psychology, Eotvos University, Szombathely, Hungary
| | - Szablics Peter
- Institute of Physical Education and Sport Science, JGYPK, University of Szeged, Szeged, Hungary
| | - Geza Toth
- Affidea Diagnostic Center, Budapest, Hungary
| | - Hans Degens
- School of Healthcare Science, Manchester Metropolitan University, UK; Institute of Sport Science and Innovations, Lithuanian Sports University, Lithuania
| | - Jörn Rittweger
- Division Space Physiology, Institute of Aerospace Medicine, German Aerospace Center, Cologne, Germany
| | - Istvan Boldogh
- University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | - Zsolt Radak
- Research Institute of Sport Science, University of Physical Education, Budapest, Hungary.
| |
Collapse
|
47
|
Botteri G, Salvadó L, Gumà A, Lee Hamilton D, Meakin PJ, Montagut G, Ashford MLJ, Ceperuelo-Mallafré V, Fernández-Veledo S, Vendrell J, Calderón-Dominguez M, Serra D, Herrero L, Pizarro J, Barroso E, Palomer X, Vázquez-Carrera M. The BACE1 product sAPPβ induces ER stress and inflammation and impairs insulin signaling. Metabolism 2018. [PMID: 29526536 DOI: 10.1016/j.metabol.2018.03.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE β-secretase/β-site amyloid precursor protein (APP)-cleaving enzyme 1 (BACE1) is a key enzyme involved in Alzheimer's disease that has recently been implicated in insulin-independent glucose uptake in myotubes. However, it is presently unknown whether BACE1 and the product of its activity, soluble APPβ (sAPPβ), contribute to lipid-induced inflammation and insulin resistance in skeletal muscle cells. MATERIALS/METHODS Studies were conducted in mouse C2C12 myotubes, skeletal muscle from Bace1-/-mice and mice treated with sAPPβ and adipose tissue and plasma from obese and type 2 diabetic patients. RESULTS We show that BACE1 inhibition or knockdown attenuates palmitate-induced endoplasmic reticulum (ER) stress, inflammation, and insulin resistance and prevents the reduction in Peroxisome Proliferator-Activated Receptor γ Co-activator 1α (PGC-1α) and fatty acid oxidation caused by palmitate in myotubes. The effects of palmitate on ER stress, inflammation, insulin resistance, PGC-1α down-regulation, and fatty acid oxidation were mimicked by soluble APPβ in vitro. BACE1 expression was increased in subcutaneous adipose tissue of obese and type 2 diabetic patients and this was accompanied by a decrease in PGC-1α mRNA levels and by an increase in sAPPβ plasma levels of obese type 2 diabetic patients compared to obese non-diabetic subjects. Acute sAPPβ administration to mice reduced PGC-1α levels and increased inflammation in skeletal muscle and decreased insulin sensitivity. CONCLUSIONS Collectively, these findings indicate that the BACE1 product sAPPβ is a key determinant in ER stress, inflammation and insulin resistance in skeletal muscle and gluconeogenesis in liver.
Collapse
Affiliation(s)
- Gaia Botteri
- Pharmacology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Laia Salvadó
- Pharmacology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Anna Gumà
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | - D Lee Hamilton
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Paul J Meakin
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Gemma Montagut
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Michael L J Ashford
- Division of Molecular and Clinical Medicine, School of Medicine, Ninewells Hospital & Medical School, University of Dundee, Dundee, UK
| | - Victoria Ceperuelo-Mallafré
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
| | - Sonia Fernández-Veledo
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
| | - Joan Vendrell
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Hospital Universitari de Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili, Universitat Rovira i Virgili, Tarragona, Spain
| | - María Calderón-Dominguez
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Barcelona, Spain
| | - Dolors Serra
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Barcelona, Spain
| | - Laura Herrero
- Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBEROBN), Instituto de Salud Carlos III, Barcelona, Spain
| | - Javier Pizarro
- Pharmacology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Emma Barroso
- Pharmacology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Xavier Palomer
- Pharmacology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain
| | - Manuel Vázquez-Carrera
- Pharmacology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, Spain; Institut de Biomedicina de la Universitat de Barcelona (IBUB), Spain; Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IR-SJD), Esplugues de Llobregat, Barcelona, Spain.
| |
Collapse
|
48
|
Henriksen TI, Heywood SE, Hansen NS, Pedersen BK, Scheele CC, Nielsen S. Single Cell Analysis Identifies the miRNA Expression Profile of a Subpopulation of Muscle Precursor Cells Unique to Humans With Type 2 Diabetes. Front Physiol 2018; 9:883. [PMID: 30050458 PMCID: PMC6050405 DOI: 10.3389/fphys.2018.00883] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/19/2018] [Indexed: 12/13/2022] Open
Abstract
MicroRNAs (miRNAs) take part in regulating central cellular processes such as differentiation and metabolism. We have previously shown that muscle progenitor cells derived from individuals with type 2 diabetes (T2DM) have a dysregulated miRNA profile. We hypothesized that the T2DM muscle progenitor population is heterogeneous in its miRNA expression and differs from the progenitor population of healthy controls. MiRNA expression profiles of CD56+ muscle progenitor cells from people with T2DM and from healthy controls were therefore investigated at a single cell level. Single-cell analysis revealed three subpopulations expressing distinct miRNA profiles: two subpopulations including both T2DM and healthy control muscle precursors presented miRNA expression profiles mostly overlapping between groups. A distinct third subpopulation consisted solely of cells from donors with T2DM and showed enriched expression of miRNAs previously shown to be associated with type 2 diabetes. Among the enriched miRNAs was miR-29, a regulator of GLUT4 mRNA expression. Interestingly, this subpopulation also revealed several miRNAs with predicted targets in the PI3K/Akt pathway, not previously described in relation to T2DM muscle dysfunction. We concluded that a subpopulation of T2DM muscle precursor cells is severely dysregulated in terms of their miRNA expression, and accumulation of this population might thus contribute to the dysfunctional muscular phenotype in type 2 diabetes.
Collapse
Affiliation(s)
- Tora I Henriksen
- Centre for Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sarah E Heywood
- Centre for Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Ninna S Hansen
- Centre for Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Bente K Pedersen
- Centre for Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Camilla C Scheele
- Centre for Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.,The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Søren Nielsen
- Centre for Inflammation and Metabolism and the Centre for Physical Activity Research, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
49
|
Mangine GT, Gonzalez AM, Townsend JR, Wells AJ, Beyer KS, Miramonti AA, Ratamess NA, Stout JR, Hoffman JR. Influence of Baseline Muscle Strength and Size Measures on Training Adaptations in Resistance-trained Men. INTERNATIONAL JOURNAL OF EXERCISE SCIENCE 2018; 11:198-213. [PMID: 29795731 PMCID: PMC5955287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The influence of baseline strength or muscle size on adaptations to training is not well-understood. Comparisons between novice and advanced lifters, and between stronger and weaker experienced-lifters, have produced conflicting results. This study examined the effect of baseline muscle strength and size on subsequent adaptations in resistance-trained individuals following a traditional high-volume, short-rest resistance training protocol. Fourteen resistance-trained men (24.0±2.7 y; 90.1±11.7 kg; 169.9±29.0 cm) completed pre-training (PRE) ultrasound measurements of muscle cross-sectional area (CSA) in the rectus femoris (RF), vastus lateralis (VL), pectoralis major, and triceps brachii (TRI) prior to strength assessments (e.g., one-repetition maximum strength bench press and back-squat). Post-training (POST) assessments were completed following 8-wks (4 d·wk-1) of resistance training. Comparisons were made between stronger (STR) and weaker (WKR) participants, and between larger (LGR) and smaller (SMR) participants, based upon PRE-muscle strength and size, respectively. When groups were based on upper-body strength, repeated measures analysis of variance indicated a significant group × time interaction where greater improvements in bench press strength were observed in WKR (12.5±8.6%, p = 0.013) compared to STR (1.3±5.4%, p=0.546). Within this comparison, STR also possessed more resistance training experience than WKR (mean difference=3.1 y, p=0.002). No other differences in experience or adaptations to training were observed. These data suggest that following a short-duration training program (8-weeks), baseline size and strength have little impact on performance gains in resistance-trained individuals who possess similar years of experience. However, when training experience is different, baseline strength may affect adaptations.
Collapse
Affiliation(s)
- Gerald T Mangine
- Exercise Science and Sport Management, Kennesaw State University, Kennesaw, GA, USA
| | | | - Jeremy R Townsend
- Exercise and Nutrition Science, Lipscomb University, Nashville, TN, USA
| | - Adam J Wells
- Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, FL, USA
| | - Kyle S Beyer
- Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, FL, USA
| | - Amelia A Miramonti
- Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, FL, USA
| | | | - Jeffrey R Stout
- Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, FL, USA
| | - Jay R Hoffman
- Institute of Exercise Physiology and Wellness, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
50
|
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
|