1
|
Alur V, Vastrad B, Raju V, Vastrad C, Kotturshetti S. The identification of key genes and pathways in polycystic ovary syndrome by bioinformatics analysis of next-generation sequencing data. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2024; 29:53. [DOI: 10.1186/s43043-024-00212-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Abstract
Background
Polycystic ovary syndrome (PCOS) is a reproductive endocrine disorder. The specific molecular mechanism of PCOS remains unclear. The aim of this study was to apply a bioinformatics approach to reveal related pathways or genes involved in the development of PCOS.
Methods
The next-generation sequencing (NGS) dataset GSE199225 was downloaded from the gene expression omnibus (GEO) database and NGS dataset analyzed is obtained from in vitro culture of PCOS patients’ muscle cells and muscle cells of healthy lean control women. Differentially expressed gene (DEG) analysis was performed using DESeq2. The g:Profiler was utilized to analyze the gene ontology (GO) and REACTOME pathways of the differentially expressed genes. A protein–protein interaction (PPI) network was constructed and module analysis was performed using HiPPIE and cytoscape. The miRNA-hub gene regulatory network and TF-hub gene regulatory network were constructed. The hub genes were validated by using receiver operating characteristic (ROC) curve analysis.
Results
We have identified 957 DEG in total, including 478 upregulated genes and 479 downregulated gene. GO terms and REACTOME pathways illustrated that DEG were significantly enriched in regulation of molecular function, developmental process, interferon signaling and platelet activation, signaling, and aggregation. The top 5 upregulated hub genes including HSPA5, PLK1, RIN3, DBN1, and CCDC85B and top 5 downregulated hub genes including DISC1, AR, MTUS2, LYN, and TCF4 might be associated with PCOS. The hub gens of HSPA5 and KMT2A, together with corresponding predicted miRNAs (e.g., hsa-mir-34b-5p and hsa-mir-378a-5p), and HSPA5 and TCF4 together with corresponding predicted TF (e.g., RCOR3 and TEAD4) were found to be significantly correlated with PCOS.
Conclusions
These study uses of bioinformatics analysis of NGS data to obtain hub genes and key signaling pathways related to PCOS and its associated complications. Also provides novel ideas for finding biomarkers and treatment methods for PCOS and its associated complications.
Collapse
|
2
|
Vastrad B, Vastrad C. Screening and identification of key biomarkers associated with endometriosis using bioinformatics and next-generation sequencing data analysis. EGYPTIAN JOURNAL OF MEDICAL HUMAN GENETICS 2024; 25:116. [DOI: 10.1186/s43042-024-00572-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/23/2024] [Indexed: 01/04/2025] Open
Abstract
Abstract
Background
Endometriosis is a common cause of endometrial-type mucosa outside the uterine cavity with symptoms such as painful periods, chronic pelvic pain, pain with intercourse and infertility. However, the early diagnosis of endometriosis is still restricted. The purpose of this investigation is to identify and validate the key biomarkers of endometriosis.
Methods
Next-generation sequencing dataset GSE243039 was obtained from the Gene Expression Omnibus database, and differentially expressed genes (DEGs) between endometriosis and normal control samples were identified. After screening of DEGs, gene ontology (GO) and REACTOME pathway enrichment analyses were performed. Furthermore, a protein–protein interaction (PPI) network was constructed and modules were analyzed using the Human Integrated Protein–Protein Interaction rEference database and Cytoscape software, and hub genes were identified. Subsequently, a network between miRNAs and hub genes, and network between TFs and hub genes were constructed using the miRNet and NetworkAnalyst tool, and possible key miRNAs and TFs were predicted. Finally, receiver operating characteristic curve analysis was used to validate the hub genes.
Results
A total of 958 DEGs, including 479 upregulated genes and 479 downregulated genes, were screened between endometriosis and normal control samples. GO and REACTOME pathway enrichment analyses of the 958 DEGs showed that they were mainly involved in multicellular organismal process, developmental process, signaling by GPCR and muscle contraction. Further analysis of the PPI network and modules identified 10 hub genes, including vcam1, snca, prkcb, adrb2, foxq1, mdfi, actbl2, prkd1, dapk1 and actc1. Possible target miRNAs, including hsa-mir-3143 and hsa-mir-2110, and target TFs, including tcf3 (transcription factor 3) and clock (clock circadian regulator), were predicted by constructing a miRNA-hub gene regulatory network and TF-hub gene regulatory network.
Conclusions
This investigation used bioinformatics techniques to explore the potential and novel biomarkers. These biomarkers might provide new ideas and methods for the early diagnosis, treatment and monitoring of endometriosis.
Collapse
|
3
|
Sabaratnam R, Hansen DR, Svenningsen P. White adipose tissue mitochondrial bioenergetics in metabolic diseases. Rev Endocr Metab Disord 2023; 24:1121-1133. [PMID: 37558853 DOI: 10.1007/s11154-023-09827-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/19/2023] [Indexed: 08/11/2023]
Abstract
White adipose tissue (WAT) is an important endocrine organ that regulates systemic energy metabolism. In metabolically unhealthy obesity, adipocytes become dysfunctional through hypertrophic mechanisms associated with a reduced endocrine function, reduced mitochondrial function, but increased inflammation, fibrosis, and extracellular remodelling. A pathologic WAT remodelling promotes systemic lipotoxicity characterized by fat accumulation in tissues such as muscle and liver, leading to systemic insulin resistance and type 2 diabetes. Several lines of evidence from human and animal studies suggest a link between unhealthy obesity and adipocyte mitochondrial dysfunction, and interventions that improve mitochondrial function may reduce the risk of obesity-associated diseases. This review discusses the importance of mitochondrial function and metabolism in human adipocyte biology and intercellular communication mechanisms within WAT. Moreover, a selected interventional approach for better adipocyte mitochondrial metabolism in humans is reviewed. A greater understanding of mitochondrial bioenergetics in WAT might provide novel therapeutic opportunities to prevent or restore dysfunctional adipose tissue in obesity-associated diseases.
Collapse
Affiliation(s)
- Rugivan Sabaratnam
- Department of Clinical Research, University of Southern Denmark, Odense C, DK-5000, Denmark.
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark.
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| | - Didde Riisager Hansen
- Steno Diabetes Center Odense, Odense University Hospital, Odense C, DK-5000, Denmark
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark
| | - Per Svenningsen
- Department of Molecular Medicine, Cardiovascular and Renal Research, University of Southern Denmark, J. B. Winsløws Vej 21,3, Odense C, DK-5000, Denmark.
| |
Collapse
|
4
|
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
|
5
|
Peroxisome Proliferator-Activated Receptors and Caloric Restriction-Common Pathways Affecting Metabolism, Health, and Longevity. Cells 2020; 9:cells9071708. [PMID: 32708786 PMCID: PMC7407644 DOI: 10.3390/cells9071708] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/14/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Caloric restriction (CR) is a traditional but scientifically verified approach to promoting health and increasing lifespan. CR exerts its effects through multiple molecular pathways that trigger major metabolic adaptations. It influences key nutrient and energy-sensing pathways including mammalian target of rapamycin, Sirtuin 1, AMP-activated protein kinase, and insulin signaling, ultimately resulting in reductions in basic metabolic rate, inflammation, and oxidative stress, as well as increased autophagy and mitochondrial efficiency. CR shares multiple overlapping pathways with peroxisome proliferator-activated receptors (PPARs), particularly in energy metabolism and inflammation. Consequently, several lines of evidence suggest that PPARs might be indispensable for beneficial outcomes related to CR. In this review, we present the available evidence for the interconnection between CR and PPARs, highlighting their shared pathways and analyzing their interaction. We also discuss the possible contributions of PPARs to the effects of CR on whole organism outcomes.
Collapse
|
6
|
Liu Y, Bi X, Zhang Y, Wang Y, Ding W. Mitochondrial dysfunction/NLRP3 inflammasome axis contributes to angiotensin II-induced skeletal muscle wasting via PPAR-γ. J Transl Med 2020; 100:712-726. [PMID: 31857693 DOI: 10.1038/s41374-019-0355-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 10/19/2019] [Accepted: 11/04/2019] [Indexed: 12/15/2022] Open
Abstract
Angiotensin II (Ang II) levels are elevated in patients with chronic kidney disease or heart failure, and directly causes skeletal muscle wasting in rodents, but the molecular mechanisms of Ang II-induced skeletal muscle wasting and its potential as a therapeutic target are unknown. We investigated the NLR family pyrin domain containing 3 (NLRP3) inflammasome-mediated muscle atrophy response to Ang II in C2C12 myotubes and Nlrp3 knockout mice. We also assessed the mitochondrial dysfunction (MtD)/NLRP3 inflammasome axis in Ang II-induced C2C12 myotubes. Finally, we examined whether a peroxisome proliferator-activated receptor-γ (PPAR-γ) agonist could attenuate skeletal muscle wasting by targeting the MtD/NLRP3 inflammasome axis in vitro and in vivo. We demonstrated that Ang II increased NLRP3 inflammasome activation in cultured C2C12 myotubes dose dependently. Nlrp3 knockdown or Nlrp3-/- mice were protected from the imbalance of protein synthesis and degradation. Exposure of C2C12 to Ang II increased mitochondrial ROS (mtROS) generation, accompanied by MtD. Remarkably, the mitochondrial-targeted antioxidant not only decreased mtROS and MtD, it also significantly inhibited NLRP3 inflammasome activation and restored skeletal muscle atrophy. Finally, the PPAR-γ agonist protected against Ang II-induced muscle wasting by preventing MtD, oxidative stress, and NLRP3 inflammasome activation in vitro and in vivo. This work suggests a potential role of MtD/NLRP3 inflammasome pathway in the pathogenesis of Ang II-induced skeletal muscle wasting, and targeting the PPAR-γ/MtD/NLRP3 inflammasome axis may provide a therapeutic approach for muscle wasting.
Collapse
Affiliation(s)
- Yuqing Liu
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Xiao Bi
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Yumei Zhang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Yingdeng Wang
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China
| | - Wei Ding
- Division of Nephrology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiaotong University, 639 zhizaoju Road, 200011, Shanghai, China.
| |
Collapse
|
7
|
Zhang X, Wang Y, Zuo F, Su Y, Liu H, Wu B, Nian X. Effect of Insulin-Regulated FOXC2 Expression in Adipocyte Differentiation and Insulin Resistance. Diabetes Metab Syndr Obes 2020; 13:2801-2809. [PMID: 32821143 PMCID: PMC7418171 DOI: 10.2147/dmso.s262950] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 07/24/2020] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE 1) To investigate the effect of FOXC2 on the differentiation of adipose-derived mesenchymal stem cells. 2) To analyze the mechanism between FOXC2 expression regulation in adipose differentiation and insulin resistance (IR). METHODS We first amplified the FOXC2 promoter region-512 and cloned it into the luciferase expression vector. The reporter gene system was transfected into the adipose tissue-derived mesenchymal stem cell to study insulin-mediated FOXC2 expression. We also manipulated FOXC2 protein expression by either siRNA or overexpression and studied the differentiation capability of adipose tissue-derived mesenchymal stem cell into adipocytes, as well as the influence on several IR-related genes: GLUT4, PPARγ, UCP1 and PAI-1. RESULTS 1) Insulin effectively induced the expression of FOXC2 protein in adipose tissue-derived mesenchymal stem cells under differentiation (P<0.01). Insulin also induced FOXC2-pro-512T promoter activity significantly (P<0.01). 2) The stem cell adipose differentiation decreased in the FOXC2 overexpression group. 3) When FOXC2 was overexpressed, the expression of GLUT4, PAI-1 and UCP1 was higher than control groups (p<0.001). When FOXC2 was down-regulated by siRNA, both GLUT4 and PAI-1's protein expression were decreased (p<0.001), and the protein expression of PPARγ was increased (p<0.001). In the presence of insulin induction, overexpression of FOXC2 led to significantly higher UCP-1 expression (p<0.001) and lower PAI-1 expression (p<0.001). The protein expression of GLUT4, PAI-1 (p<0.001) and UCP-1 (p<0.05) was decreased in cells transfected with FOXC2 siRNA. CONCLUSION Insulin effectively induced the expression of FOXC2 protein in adipose tissue-derived mesenchymal stem cells under differentiation, possibly through the regulation of the FOXC2-pro-512T promoter activity. The different protein expression of FOXC2 has regulatory effects on several genes related to insulin resistance. FOXC2 is an important regulatory factor in adipocyte differentiation and insulin resistance.
Collapse
Affiliation(s)
- Xuxiang Zhang
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Yuming Wang
- Department of Clinical Laboratory, Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Fan Zuo
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Yandan Su
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Hua Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Bin Wu
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
| | - Xin Nian
- Department of Endocrinology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, People’s Republic of China
- Correspondence: Xin NianDepartment of Endocrinology, First Affiliated Hospital of Kunming Medical University Email
| |
Collapse
|
8
|
Ramirez-Peña E, Arnold J, Shivakumar V, Joseph R, Vidhya Vijay G, den Hollander P, Bhangre N, Allegakoen P, Prasad R, Conley Z, Matés JM, Márquez J, Chang JT, Vasaikar S, Soundararajan R, Sreekumar A, Mani SA. The Epithelial to Mesenchymal Transition Promotes Glutamine Independence by Suppressing GLS2 Expression. Cancers (Basel) 2019; 11:cancers11101610. [PMID: 31652551 PMCID: PMC6826439 DOI: 10.3390/cancers11101610] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 10/05/2019] [Accepted: 10/18/2019] [Indexed: 12/17/2022] Open
Abstract
Identifying bioenergetics that facilitate the epithelial to mesenchymal transition (EMT) in breast cancer cells may uncover targets to treat incurable metastatic disease. Metastasis is the number one cause of cancer-related deaths; therefore, it is urgent to identify new treatment strategies to prevent the initiation of metastasis. To characterize the bioenergetics of EMT, we compared metabolic activities and gene expression in cells induced to differentiate into the mesenchymal state with their epithelial counterparts. We found that levels of GLS2, which encodes a glutaminase, are inversely associated with EMT. GLS2 down-regulation was correlated with reduced mitochondrial activity and glutamine independence even in low-glucose conditions. Restoration of GLS2 expression in GLS2-negative breast cancer cells rescued mitochondrial activity, enhanced glutamine utilization, and inhibited stem-cell properties. Additionally, inhibition of expression of the transcription factor FOXC2, a critical regulator of EMT in GLS2-negative cells, restored GLS2 expression and glutamine utilization. Furthermore, in breast cancer patients, high GLS2 expression is associated with improved survival. These findings suggest that epithelial cancer cells rely on glutamine and that cells induced to undergo EMT become glutamine independent. Moreover, the inhibition of EMT leads to a GLS2-directed metabolic shift in mesenchymal cancer cells, which may make these cells susceptible to chemotherapies.
Collapse
Affiliation(s)
- Esmeralda Ramirez-Peña
- National Cancer Institute, Cancer Prevention Fellowship Program, Division of Cancer Prevention, Bethesda, MD 20892, USA.
| | - James Arnold
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Vinita Shivakumar
- Wiess School of Natural Sciences, Rice University, Houston, TX 77005, USA.
| | - Robiya Joseph
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | | | - Petra den Hollander
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Neeraja Bhangre
- Department of Fibrosis Biology, Gilead Sciences, Foster City, CA 94404, USA.
| | - Paul Allegakoen
- Department of Medicine, University of California-San Francisco, San Francisco, CA 94143, USA.
| | - Rishika Prasad
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Zachary Conley
- Center for Science Outreach, Department of Teaching and Learning, Vanderbilt University, Nashville, TN 37235, USA.
| | - José M Matés
- Canceromics Lab, Department of Molecular Biology and Biochemistry, University of Málaga and Instituto de Investigación Biomedica de Málaga (IBIMA), 29071 Málaga, Spain.
| | - Javier Márquez
- Canceromics Lab, Department of Molecular Biology and Biochemistry, University of Málaga and Instituto de Investigación Biomedica de Málaga (IBIMA), 29071 Málaga, Spain.
| | - Jeffrey T Chang
- Department of Integrative Biology and Pharmacology, University of Texas Health Science Center, Houston, TX 77030, USA.
| | - Suhas Vasaikar
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| | - Arun Sreekumar
- Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| | - Sendurai A Mani
- Department of Translational Molecular Pathology, MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Han S, Yang H, Han Y, Zhang H. Genes and transcription factors related to the adverse effects of maternal type I diabetes mellitus on fetal development. Mol Cell Probes 2018; 43:64-71. [PMID: 30447278 DOI: 10.1016/j.mcp.2018.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 10/15/2018] [Accepted: 11/13/2018] [Indexed: 11/24/2022]
Abstract
PURPOSE Maternal type I diabetes mellitus (T1DM) increases the risk of adverse pregnancy outcomes, but the corresponding mechanism is unclear. This study aims to investigate the mechanism underlying the adverse pregnancy outcomes of maternal T1DM. METHODS Gene expression microarray (GSE51546) was down-loaded from the Gene Expression Omnibus. This dataset included 12 umbilical cord samples from the newborns of T1DM mothers (T1DM group, N = six) and non-diabetic mothers (control group, N = six). RESULTS Consequently, 1051 differentially expressed genes (DEGs) were found between the two groups. The up-regulated DEGs enriched in 30 KEGG pathways. HLA-DPA1, HLA-DMA, HLA-DMB, HLA-DQA1, HLA-DQA2 and HLA-DRA enriched in "Type I diabetes mellitus". This pathway was strongly related to 14 pathways, most of which were associated with diseases. Then, a protein-protein interaction network was constructed, and 45 potential key DEGs were identified. The 45 DEGs enriched in pathways such as "Rheumatoid arthritis", "Chemokine signaling pathway" and "Cytokine-cytokine receptor interaction" (e.g. CXCL12 and CCL5). Transcription factors (TFs) of key DEGs were predicted, and a TF-DEG regulatory network was constructed. CONCLUSIONS Some genes (e.g. CXCL12 and CCL5) and their TFs were significantly and abnormally regulated in the umbilical cord tissue from the pregnancies of T1DM mothers compared to that from non-T1DM mothers.
Collapse
Affiliation(s)
- Shuyi Han
- Department of Clinical Laboratory, Ji'nan Central Hospital Affiliated to Shandong University, Ji'nan, 250013, China
| | - Huili Yang
- Department of Obstetrics, Ji'nan Central Hospital Affiliated to Shandong University, Ji'nan, 250013, China.
| | - Yunhui Han
- Department of Obstetrics, Ji'nan Central Hospital Affiliated to Shandong University, Ji'nan, 250013, China
| | - Hongzhi Zhang
- Department of Gynecology, Ji'nan Central Hospital Affiliated to Shandong University, Ji'nan, 250013, China
| |
Collapse
|
10
|
Yu JC, Hsu FC, Chiu YF. Assessment of fenofibrate-methylation interactions on triglycerides using longitudinal family data. BMC Proc 2018; 12:48. [PMID: 30263049 PMCID: PMC6156834 DOI: 10.1186/s12919-018-0132-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Background Triglyceride (TG) concentrations decrease in response to fenofibrate treatment, and also are associated with DNA methylation. But how interactions between fenofibrate response and DNA methylation affect TGs remains unclear. Methods In the present study, we identified and compared differential methylation sites associated with TG concentrations in individuals before and after fenofibrate treatment. We then estimated interactions between fenofibrate treatment and methylation to identify differential methylation effects associated with fenofibrate treatment on TG concentrations using the entire longitudinal family sample. To account for within-family and within-individual corrections, the generalized estimating equations approach was used to estimate main and interaction effects between methylation sites and fenofibrate treatment, adjusting for potential confounders. Analyses were also performed with and without adjusting for high-density lipoprotein (HDL) concentrations. Results Prior to fenofibrate treatment, 23 cytosine-phosphate-guanine (CpG) sites were significantly associated with TG concentrations, while only 13 CpG sites were identified posttreatment, adjusting for HDL. Without adjusting for HDL, pretreatment, 20 CpG sites were significantly associated with TG concentrations, while only 12 CpG sites were identified posttreatment. Among these sites, only one differential site (cg19003390 in the CPT1A gene) overlapped from pre- and posttreatment measurements regardless of HDL adjustment. Furthermore, 11 methylation sites showed substantial interaction effects (p < 1.43 × 10−7with Bonferroni correction) with or without HDL adjustment when using the whole longitudinal data. Conclusions Our analyses suggest that DNA methylation likely modified the effect of fenofibrate on TG concentrations. Differential fenofibrate-associated methylation sites on TGs differed with and without adjusting for HDL concentrations, suggesting that these HDLs and TGs might share some common epigenetic processes.
Collapse
Affiliation(s)
- Jih-Chang Yu
- 1Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053 Taiwan
| | - Fang-Chi Hsu
- 2Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest School of Medicine, Medical City Boulevard, Winston-Salem, NC 27157 USA
| | - Yen-Feng Chiu
- 1Institute of Population Health Sciences, National Health Research Institutes, 35 Keyan Road, Zhunan, Miaoli, 35053 Taiwan
| |
Collapse
|
11
|
Lee TW, Bai KJ, Lee TI, Chao TF, Kao YH, Chen YJ. PPARs modulate cardiac metabolism and mitochondrial function in diabetes. J Biomed Sci 2017; 24:5. [PMID: 28069019 PMCID: PMC5223385 DOI: 10.1186/s12929-016-0309-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 12/05/2016] [Indexed: 01/08/2023] Open
Abstract
Diabetic cardiomyopathy is a major complication of diabetes mellitus (DM). Currently, effective treatments for diabetic cardiomyopathy are limited. The pathophysiology of diabetic cardiomyopathy is complex, whereas mitochondrial dysfunction plays a vital role in the genesis of diabetic cardiomyopathy. Metabolic regulation targeting mitochondrial dysfunction is expected to be a reasonable strategy for treating diabetic cardiomyopathy. Peroxisome proliferator-activated receptors (PPARs) are master executors in regulating glucose and lipid homeostasis and also modulate mitochondrial function. However, synthetic PPAR agonists used for treating hyperlipidemia and DM have shown controversial effects on cardiovascular regulation. This article reviews our updated understanding of the beneficial and detrimental effects of PPARs on mitochondria in diabetic hearts.
Collapse
Affiliation(s)
- Ting-Wei Lee
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.,Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Jen Bai
- Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,School of Respiratory Therapy, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ting-I Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,Department of General Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tze-Fan Chao
- Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan.,Institute of Clinical Medicine, and Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Hsun Kao
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan. .,Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, 250 Wu-Xing Street, Taipei, 11031, Taiwan.,Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
12
|
Goltsman I, Khoury EE, Winaver J, Abassi Z. Does Thiazolidinedione therapy exacerbate fluid retention in congestive heart failure? Pharmacol Ther 2016; 168:75-97. [PMID: 27598860 DOI: 10.1016/j.pharmthera.2016.09.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The ever-growing global burden of congestive heart failure (CHF) and type 2 diabetes mellitus (T2DM) as well as their co-existence necessitate that anti-diabetic pharmacotherapy will modulate the cardiovascular risk inherent to T2DM while complying with the accompanying restrictions imposed by CHF. The thiazolidinedione (TZD) family of peroxisome proliferator-activated receptor γ (PPARγ) agonists initially provided a promising therapeutic option in T2DM owing to anti-diabetic efficacy combined with pleiotropic beneficial cardiovascular effects. However, the utility of TZDs in T2DM has declined in the past decade, largely due to concomitant adverse effects of fluid retention and edema formation attributed to salt-retaining effects of PPARγ activation on the nephron. Presumably, the latter effects are potentially deleterious in the context of pre-existing fluid retention in CHF. However, despite a considerable body of evidence on mechanisms responsible for TZD-induced fluid retention suggesting that this class of drugs is rightfully prohibited from use in CHF patients, there is a paucity of experimental and clinical studies that investigate the effects of TZDs on salt and water homeostasis in the CHF setting. In an attempt to elucidate whether TZDs actually exacerbate the pre-existing fluid retention in CHF, our review summarizes the pathophysiology of fluid retention in CHF. Moreover, we thoroughly review the available data on TZD-induced fluid retention and proposed mechanisms in animals and patients. Finally, we will present recent studies challenging the common notion that TZDs worsen renal salt and water retention in CHF.
Collapse
Affiliation(s)
- Ilia Goltsman
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Emad E Khoury
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Joseph Winaver
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology, Biophysics and Systems Biology, The Bruce Rappaport, Rappaport Faculty of Medicine, Technion, Haifa, Israel; Department of Laboratory Medicine, Rambam Human Health Care Campus, Haifa, Israel.
| |
Collapse
|
13
|
Das NR, Sharma SS. Cognitive Impairment Associated with Parkinson's Disease: Role of Mitochondria. Curr Neuropharmacol 2016; 14:584-92. [PMID: 26725887 PMCID: PMC4981741 DOI: 10.2174/1570159x14666160104142349] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 08/27/2015] [Accepted: 01/01/2016] [Indexed: 11/22/2022] Open
Abstract
Parkinson's disease (PD) is a movement disorder and is associated with some of the intellectual disabilities like cognitive dysfunctions. PD associated cognitive dysfunctions have been proved well in both preclinical and clinical set ups. Like other neurodegenerative diseases, insults to mitochondria have a significant role in the pathobiology of PD associated dementia (PDD). Neurotoxins like MPTP, mutations of the mitochondrial genes, oxidative stress, imbalanced redox mechanisms and dysregulated mitochondrial dynamics have been implicated in mitochondrial dysfunctions and have paramount importance in the pathobiology of PDD. However, the extent of contribution of mitochondrial dysfunctions towards cognitive deficits in PD has not been characterized completely. In this review we highlight on the contribution of mitochondrial dysfunction to PDD. We also highlight different behavioural tests used in nonhuman primate and rodent models for assessing cognitive deficits and some common techniques for evaluation of mitochondrial dysfunction in PDD.
Collapse
Affiliation(s)
| | - Shyam S Sharma
- Molecular Neuropharmacology Laboratory, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector-67, SAS Nagar, Punjab-160062, India.
| |
Collapse
|
14
|
Datta N, Lindfors S, Miura N, Saleem MA, Lehtonen S. Overexpression of transcription factor FOXC2 in cultured human podocytes upregulates injury markers and increases motility. Exp Cell Res 2015; 340:32-42. [PMID: 26524507 DOI: 10.1016/j.yexcr.2015.10.035] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 10/09/2015] [Accepted: 10/29/2015] [Indexed: 01/23/2023]
Abstract
Obesity and diabetes-related kidney diseases associate with renal failure and cardiovascular morbidity, and represent a major health issue worldwide. However, the molecular mechanisms leading to their development remain poorly understood. We observed increased expression of transcription factor FoxC2 in the podocytes of obese Zucker rats that are insulin resistant and albuminuric. We also found that depletion of adiponectin, an adipocyte-derived hormone whose secretion is decreased in obesity, upregulated FOXC2 in differentiated human podocytes in vitro. Overexpression of FOXC2 in cultured human podocytes led to increased nuclear expression of FOXC2 associated with a change of cellular morphology. This was accompanied by upregulation of vimentin, a key mesenchymal marker, and active beta-catenin, associated with podocyte injury. We also observed re-organization of the actin cytoskeleton, disrupted localization of the tight junction protein ZO-1, and increased motility of podocytes overexpressing FOXC2. These data indicate that the expression of FOXC2 in podocytes needs to be tightly regulated, and that its overexpression induces a chain of cellular events leading to podocyte dysfunction. These changes may lead to podocyte detachment and depletion ultimately contributing to albuminuria. We also suggest a novel molecular mechanism linking obesity-induced decrease in adiponectin to podocyte dysfunction via upregulation of FOXC2.
Collapse
Affiliation(s)
- Neeta Datta
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | - Sonja Lindfors
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland
| | - Naoyuki Miura
- Department of Biochemistry, Hamamatsu University School of Medicine, Hamamatsu 431-3192, Japan
| | - Moin A Saleem
- Academic and Children's Renal Unit, Dorothy Hodgkin Building, Bristol BS1, United Kingdom
| | - Sanna Lehtonen
- Department of Pathology, University of Helsinki, 00290 Helsinki, Finland.
| |
Collapse
|
15
|
Gan L, Liu Z, Jin W, Zhou Z, Sun C. Foxc2 enhances proliferation and inhibits apoptosis through activating Akt/mTORC1 signaling pathway in mouse preadipocytes. J Lipid Res 2015; 56:1471-80. [PMID: 26113535 DOI: 10.1194/jlr.m057679] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Indexed: 12/21/2022] Open
Abstract
Forkhead box C2 (Foxc2) protein is a transcription factor in regulation of development, metabolism, and immunology. However, the regulatory mechanisms of Foxc2 on proliferation and apoptosis of preadipocytes are unclear. In this study, we found that high-fat-diet-induced obesity elevated the expression of Foxc2 and cyclin E after 6 weeks. Additionally, Foxc2 suppressed preadipocyte differentiation, increased cell counts and augmented G1-S transition of preadipocytes, along with the elevation of cyclin E expression and the reduction levels of p27 and p53. Furthermore, Foxc2 knockdown reduced early apoptotic cells with accompanying reduction of mitochondrial membrane potential and increased fragmentation of genomic DNA. We show that Foxc2 reduces the expression of Bax, caspase-9, and caspase-3 in both serum-starved and palmitic acid-induced cell apoptotic models, which confirms the anti-apoptotic role of Foxc2. Moreover, the protein kinase B (Akt)/mammalian target of rapamycin (mTOR)C1 signaling pathway and the ERK/mTORC1 signaling pathway were activated along with preadipocyte proliferation in response to Foxc2 overexpression, whereas apoptosis marker genes were downregulated during this process. Those effects were blocked by the interference of Foxc2 or signal pathways specific inhibitors. These data collectively reveal that Foxc2 enhances proliferation of preadipocytes and inhibits apoptosis of preadipocytes by activating the Akt/mTORC1 and ERK/mTORC1 signaling pathways.
Collapse
Affiliation(s)
- Lu Gan
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhenjiang Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Wei Jin
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Zhongjie Zhou
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chao Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China
| |
Collapse
|
16
|
Demine S, Reddy N, Renard P, Raes M, Arnould T. Unraveling biochemical pathways affected by mitochondrial dysfunctions using metabolomic approaches. Metabolites 2014; 4:831-78. [PMID: 25257998 PMCID: PMC4192695 DOI: 10.3390/metabo4030831] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 09/02/2014] [Accepted: 09/18/2014] [Indexed: 02/06/2023] Open
Abstract
Mitochondrial dysfunction(s) (MDs) can be defined as alterations in the mitochondria, including mitochondrial uncoupling, mitochondrial depolarization, inhibition of the mitochondrial respiratory chain, mitochondrial network fragmentation, mitochondrial or nuclear DNA mutations and the mitochondrial accumulation of protein aggregates. All these MDs are known to alter the capacity of ATP production and are observed in several pathological states/diseases, including cancer, obesity, muscle and neurological disorders. The induction of MDs can also alter the secretion of several metabolites, reactive oxygen species production and modify several cell-signalling pathways to resolve the mitochondrial dysfunction or ultimately trigger cell death. Many metabolites, such as fatty acids and derived compounds, could be secreted into the blood stream by cells suffering from mitochondrial alterations. In this review, we summarize how a mitochondrial uncoupling can modify metabolites, the signalling pathways and transcription factors involved in this process. We describe how to identify the causes or consequences of mitochondrial dysfunction using metabolomics (liquid and gas chromatography associated with mass spectrometry analysis, NMR spectroscopy) in the obesity and insulin resistance thematic.
Collapse
Affiliation(s)
- Stéphane Demine
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Nagabushana Reddy
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Patricia Renard
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Martine Raes
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| | - Thierry Arnould
- Laboratory of Biochemistry and Cell Biology (URBC), NARILIS (Namur Research Institute for Life Sciences), University of Namur (UNamur), 61 rue de Bruxelles, Namur 5000, Belgium.
| |
Collapse
|
17
|
Rogers C, Davis B, Neufer PD, Murphy MP, Anderson EJ, Robidoux J. A transient increase in lipid peroxidation primes preadipocytes for delayed mitochondrial inner membrane permeabilization and ATP depletion during prolonged exposure to fatty acids. Free Radic Biol Med 2014; 67:330-41. [PMID: 24269897 PMCID: PMC3935619 DOI: 10.1016/j.freeradbiomed.2013.11.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 10/29/2013] [Accepted: 11/12/2013] [Indexed: 12/22/2022]
Abstract
Preadipocytes are periodically subjected to fatty acid (FA) concentrations that are potentially cytotoxic. We tested the hypothesis that prolonged exposure of preadipocytes of human origin to a physiologically relevant mix of FAs leads to mitochondrial inner membrane (MIM) permeabilization and ultimately to mitochondrial crisis. We found that exposure of preadipocytes to FAs led to progressive cyclosporin A-sensitive MIM permeabilization, which in turn caused a reduction in MIM potential, oxygen consumption, and ATP synthetic capacity and, ultimately, death. Additionally, we showed that FAs induce a transient increase in intramitochondrial reactive oxygen species (ROS) and lipid peroxide production, lasting roughly 30 and 120min for the ROS and lipid peroxides, respectively. MIM permeabilization and its deleterious consequences including mitochondrial crisis and cell death were prevented by treating the cells with the mitochondrial FA uptake inhibitor etomoxir, the mitochondrion-selective superoxide and lipid peroxide antioxidants MitoTempo and MitoQ, or the lipid peroxide and reactive carbonyl scavenger l-carnosine. FAs also promoted a delayed oxidative stress phase. However, the beneficial effects of etomoxir, MitoTempo, and l-carnosine were lost by delaying the treatment by 2h, suggesting that the initial phase was sufficient to prime the cells for the delayed MIM permeabilization and mitochondrial crisis. It also suggested that the second ROS production phase is a consequence of this loss in mitochondrial health. Altogether, our data suggest that approaches designed to diminish intramitochondrial ROS or lipid peroxide accumulation, as well as MIM permeabilization, are valid mechanism-based therapeutic avenues to prevent the loss in preadipocyte metabolic fitness associated with prolonged exposure to elevated FA levels.
Collapse
Affiliation(s)
- Carlyle Rogers
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC 27834, USA
| | - Barbara Davis
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC 27834, USA
| | - P Darrell Neufer
- Department of Physiology, East Carolina University, Greenville, NC 27834, USA; Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, Wellcome Trust/MRC Building, Cambridge CB2 0XY, UK
| | - Ethan J Anderson
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC 27834, USA; Department of Kinesiology, East Carolina University, Greenville, NC 27834, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA
| | - Jacques Robidoux
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC 27834, USA; East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC 27834, USA.
| |
Collapse
|
18
|
Bullon P, Battino M, Varela-Lopez A, Perez-Lopez P, Granados-Principal S, Ramirez-Tortosa MC, Ochoa JJ, Cordero MD, Gonzalez-Alonso A, Ramirez-Tortosa CL, Rubini C, Zizzi A, Quiles JL. Diets based on virgin olive oil or fish oil but not on sunflower oil prevent age-related alveolar bone resorption by mitochondrial-related mechanisms. PLoS One 2013; 8:e74234. [PMID: 24066124 PMCID: PMC3774624 DOI: 10.1371/journal.pone.0074234] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 07/31/2013] [Indexed: 11/18/2022] Open
Abstract
Background/Objectives Aging enhances frequency of chronic diseases like cardiovascular diseases or periodontitis. Here we reproduced an age-dependent model of the periodontium, a fully physiological approach to periodontal conditions, to evaluate the impact of dietary fat type on gingival tissue of young (6 months old) and old (24 months old) rats. Methods/Findings Animals were fed life-long on diets based on monounsaturated fatty acids (MUFA) as virgin olive oil, n-6 polyunsaturated fatty acids (n-6PUFA), as sunflower oil, or n-3PUFA, as fish oil. Age-related alveolar bone loss was higher in n-6PUFA fed rats, probably as a consequence of the ablation of the cell capacity to adapt to aging. Gene expression analysis suggests that MUFA or n-3PUFA allowed mitochondria to maintain an adequate turnover through induction of biogenesis, autophagy and the antioxidant systems, and avoiding mitochondrial electron transport system alterations. Conclusions The main finding is that the enhanced alveolar bone loss associated to age may be targeted by an appropriate dietary treatment. The mechanisms involved in this phenomenon are related with an ablation of the cell capacity to adapt to aging. Thus, MUFA or n-3PUFA might allow mitochondrial maintaining turnover through biogenesis or autophagy. They might also be able to induce the corresponding antioxidant systems to counteract age-related oxidative stress, and do not inhibit mitochondrial electron transport chain. From the nutritional and clinical point of view, it is noteworthy that the potential treatments to attenuate alveolar bone loss (a feature of periodontal disease) associated to age could be similar to some of the proposed for the prevention and treatment of cardiovascular diseases, a group of pathologies recently associated with age-related periodontitis.
Collapse
Affiliation(s)
- Pedro Bullon
- Department of Periodontology, Dental School, University of Sevilla, Sevilla, Spain
| | - Maurizio Battino
- Dipartimento di Scienze Cliniche Specialistiche ed Odontostomatologiche, Università Politecnica delle Marche, Ancona, Italia
- * E-mail:
| | - Alfonso Varela-Lopez
- Institute of Nutrition and Food Technology “José Mataix Verdú”, Department of Physiology, University of Granada, Granada, Spain
| | - Patricia Perez-Lopez
- Institute of Nutrition and Food Technology “José Mataix Verdú”, Department of Physiology, University of Granada, Granada, Spain
| | - Sergio Granados-Principal
- Institute of Nutrition and Food Technology “José Mataix Verdú”, Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
| | - Maria C. Ramirez-Tortosa
- Institute of Nutrition and Food Technology “José Mataix Verdú”, Department of Biochemistry and Molecular Biology II, University of Granada, Granada, Spain
| | - Julio J. Ochoa
- Institute of Nutrition and Food Technology “José Mataix Verdú”, Department of Physiology, University of Granada, Granada, Spain
| | - Mario D. Cordero
- Departamento de Citología e Histología Normal y Patológica, Universidad de Sevilla, Sevilla, Spain
| | - Adrian Gonzalez-Alonso
- Institute of Nutrition and Food Technology “José Mataix Verdú”, Department of Physiology, University of Granada, Granada, Spain
| | | | - Corrado Rubini
- Dipartimento di Scienze Biomediche Sanità Pubblica, Università Politecnica delle Marche, Ancona, Italia
| | - Antonio Zizzi
- Dipartimento di Scienze Biomediche Sanità Pubblica, Università Politecnica delle Marche, Ancona, Italia
| | - José L. Quiles
- Institute of Nutrition and Food Technology “José Mataix Verdú”, Department of Physiology, University of Granada, Granada, Spain
| |
Collapse
|
19
|
Moreno-Navarrete JM, Ortega F, Serrano M, Rodriguez-Hermosa JI, Ricart W, Mingrone G, Fernández-Real JM. CIDEC/FSP27 and PLIN1 gene expression run in parallel to mitochondrial genes in human adipose tissue, both increasing after weight loss. Int J Obes (Lond) 2013; 38:865-72. [PMID: 24126816 DOI: 10.1038/ijo.2013.171] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Revised: 08/20/2013] [Accepted: 08/22/2013] [Indexed: 01/05/2023]
Abstract
OBJECTIVE FSP27 KO mice showed enhanced expression of mitochondrial genes, increased mitochondrial activity and smaller lipid droplets. Here, we aimed to investigate lipid droplet protein (CIDEC/FSP27 and perilipinA (PLIN1)) gene expression in human adipose tissue in association with obesity, insulin resistance and mitochondrial gene expression. DESIGN AND SUBJECTS In cohort 1, CIDEC/FSP27, PLIN1, adipogenic (FASN, ACACA, PPARG, GLUT4) and mitochondrial (PPARGC1A, PPARGC1B, TFAM, MT-CO3) gene expression were analyzed in 171 adipose tissue samples (88 visceral adipose tissue (VAT) and 83 subcutaneous adipose tissue (SAT) depots) and in a time course experiment in human subcutaneous and visceral preadipocytes using real-time PCR. In cohort 2, the effects of bariatric surgery-induced weight loss were also evaluated in six caucasian morbidly obese women. Additionally, in cohort 2 FSP27 and PLIN1 protein levels were measured using western blotting. RESULTS CIDEC/FSP27 (1.03±0.52 vs 0.49±0.23 relative gene expression unit (R.U.), P<0.0001) and PLIN1 (1.32±0.82 vs 0.63±0.42 R.U., P<0.0001) gene were significantly more expressed in SAT than in VAT. In VAT, CIDEC/FSP27 and PLIN1 gene expression decreased with body mass index, percent fat mass, fasting glucose, fasting insulin, HOMA and were positively associated with adipogenic (PPARG, GLUT4, FASN and ACACA) and mitochondrial biogenesis (PPARGC1A, PPARGC1B, TFAM and MT-CO3)-related genes. Mitochondrial gene expression increased during adipocyte differentiation in parallel to FSP27 and PLIN1 and other adipogenic genes. After bariatric surgery-induced weight loss, PLIN1 and CIDEC/FSP27 gene and protein expression in SAT increased significantly in parallel to adipogenic and mitochondrial genes. CONCLUSION These findings suggest a positive functional interaction between CIDEC/FSP27, PLIN1 and mitochondrial biogenesis-related genes in human adipose tissue.
Collapse
Affiliation(s)
- J M Moreno-Navarrete
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - F Ortega
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - M Serrano
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - J I Rodriguez-Hermosa
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - W Ricart
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| | - G Mingrone
- Institute of Internal Medicine, Catholic University of Rome, Rome, Italy
| | - J M Fernández-Real
- Department of Diabetes, Endocrinology and Nutrition, Institut d'Investigació Biomèdica de Girona (IdIBGi), CIBEROBN (CB06/03/010) and Instituto de Salud Carlos III (ISCIII), Girona, Spain
| |
Collapse
|
20
|
Dietary obesity-induced Egr-1 in adipocytes facilitates energy storage via suppression of FOXC2. Sci Rep 2013; 3:1476. [PMID: 23502673 PMCID: PMC3600596 DOI: 10.1038/srep01476] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/28/2013] [Indexed: 01/27/2023] Open
Abstract
The molecular mechanism to regulate energy balance is not completely understood. Here we observed that Egr-1 expression in white adipose tissue (WAT) was highly correlated with dietary-induced obesity and insulin resistance both in mice and humans. Egr-1 null mice were protected from diet-induced obesity and obesity-associated pathologies such as fatty liver, insulin resistance, hyperlipidemia and hyperinsulinemia. This phenotype can be largely explained by the increase of energy expenditure in Egr-1 null mice. Characterization of these mice revealed that the expression of FOXC2 and its target genes were significantly elevated in white adipose tissues, leading to WAT energy expenditure instead of energy storage. Altogether, these studies suggest an important role for Egr-1, which, by repressing FOXC2 expression, promotes energy storage in WAT and favored the development of obesity under high energy intake.
Collapse
|
21
|
Wright DC. Exercise- and resveratrol-mediated alterations in adipose tissue metabolism. Appl Physiol Nutr Metab 2013; 39:109-16. [PMID: 24476464 DOI: 10.1139/apnm-2013-0316] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Owing to its obligatory role in locomotion and the fact that it accounts for the vast majority of whole-body glucose and lipid oxidation, much work has focused on studying the biochemical adaptations that occur in skeletal muscle in response to exercise. However, over the past several years there has been a growing appreciation that adipose tissue is an important player in regulating systemic carbohydrate and lipid homeostasis. Despite this, the examination of how exercise alters adipose tissue function and metabolism is, when compared with skeletal muscle, in its infancy. The purpose of the current review is to highlight some of the recent findings from our laboratory and others that focus on the emerging area of adipose tissue exercise biochemistry. Specifically, the role of exercise on the induction of mitochondrial and glyceroneogenic enzymes will be examined and will be compared with the well-characterized effects of thiazolidinediones, which are insulin-sensitizing drugs. A particular emphasis will be placed on the role of interleukin-6 in mediating the effects of exercise. Finally, we will discuss recent data from our laboratory demonstrating beneficial effects of resveratrol supplementation on adipose tissue metabolism.
Collapse
Affiliation(s)
- David C Wright
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph ON N1G 2W1, Canada
| |
Collapse
|
22
|
Rahman S, Lu Y, Czernik PJ, Rosen CJ, Enerback S, Lecka-Czernik B. Inducible brown adipose tissue, or beige fat, is anabolic for the skeleton. Endocrinology 2013; 154:2687-701. [PMID: 23696565 PMCID: PMC3713216 DOI: 10.1210/en.2012-2162] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is known that insulin resistance and type 2 diabetes mellitus are associated with increased fractures and that brown adipose tissue (BAT) counteracts many if not all of the symptoms associated with type 2 diabetes. By the use of FoxC2(AD)(+/Tg) mice, a well-established model for induction of BAT, or beige fat, we present data extending the beneficial action of beige fat to also include a positive effect on bone. FoxC2(AD)(+/Tg) mice are lean and insulin-sensitive and have high bone mass due to increased bone formation associated with high bone turnover. Inducible BAT is linked to activation of endosteal osteoblasts whereas osteocytes have decreased expression of the Sost transcript encoding sclerostin and elevated expression of Rankl. Conditioned media (CM) collected from forkhead box c2 (FOXC2)-induced beige adipocytes activated the osteoblast phenotype and increased levels of phospho-AKT and β-catenin in recipient cells. In osteocytes, the same media decreased Sost expression. Immunodepletion of CM with antibodies against wingless related MMTV integration site 10b (WNT10b) and insulin-like growth factor binding protein 2 (IGFBP2) resulted in the loss of pro-osteoblastic activity, and the loss of increase in the levels of phospho-AKT and β-catenin. Conversely, CM derived from cells overexpressing IGFBP2 or WNT10b restored osteoblastic activity in recipient cells. In conclusion, beige fat secretes endocrine/paracrine activity that is beneficial for the skeleton.
Collapse
Affiliation(s)
- Sima Rahman
- Departments of Orthopaedic Surgery, University ofToledo Health Sciences Campus, Toledo, OH 43614, USA
| | | | | | | | | | | |
Collapse
|
23
|
Pangare M, Makino A. Mitochondrial function in vascular endothelial cell in diabetes. J Smooth Muscle Res 2012; 48:1-26. [PMID: 22504486 DOI: 10.1540/jsmr.48.1] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Micro- and macrovascular complications are commonly seen in diabetic patients and endothelial dysfunction contributes to the development and progression of the complications. Abnormal functions in endothelial cells lead to the increase in vascular tension and atherosclerosis, followed by systemic hypertension as well as increased incidence of ischemia and stroke in diabetic patients. Mitochondria are organelles serving as a source of energy production and as regulators of cell survival (e.g., apoptosis and cell development) and ion homeostasis (e.g., H(+), Ca(2+)). Endothelial mitochondria are mainly responsible for generation of reactive oxygen species (ROS) and maintaining the Ca(2+) concentration in the cytosol. There is increasing evidence that mitochondrial morphological and functional changes are implicated in vascular endothelial dysfunction. Enhanced mitochondrial fission and/or attenuated fusion lead to mitochondrial fragmentation and disrupt the endothelial physiological function. Abnormal mitochondrial biogenesis and disturbance of mitochondrial autophagy increase the accumulation of damaged mitochondria, such as irreversibly depolarized or leaky mitochondria, and facilitate cell death. Augmented mitochondrial ROS production and Ca(2+) overload in mitochondria not only cause the maladaptive effect on the endothelial function, but also are potentially detrimental to cell survival. In this article, we review the physiological and pathophysiological role of mitochondria in endothelial function with special focus on diabetes.
Collapse
Affiliation(s)
- Meenal Pangare
- University of Illinois at Chicago, Chicago, IL 60612, USA
| | | |
Collapse
|