1
|
Zhang M, Sha Y, Wang J, Qi H, Shi P, Liu Y, Jiang M, Ba L, Liu Y, Cao Y, Zhang Q, Sun H. Inhibition of ULK1 attenuates ferroptosis-mediated cardiac hypertrophy via HMGA2/METTL14/SLC7A11 axis in mice. Eur J Pharmacol 2025; 995:177416. [PMID: 39993699 DOI: 10.1016/j.ejphar.2025.177416] [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: 06/26/2024] [Revised: 01/24/2025] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
UNC-51-like kinase 1 (ULK1), a primary serine/threonine kinase, is implicated in diverse pathophysiological processes. Previous findings have linked ULK1-dependent autophagy to cardiac hypertrophy. Our study further explored the functional role and molecular mechanisms of ULK1 in non-autophagic signaling pathways. Notably, ULK1 expression was significantly elevated in both transverse aortic constriction (TAC)-induced hypertrophic mouse hearts and Angiotensin II (Ang II)-treated cardiomyocytes, suggesting an increased sensitivity to hypertrophic stimuli potentially mediated by ULK1-induced ferroptosis in hypertrophic cardiomyocytes. Treatment with the ferroptosis inhibitor ferrostatin-1 (Fer-1) effectively reduced ULK1-induced cardiomyocyte hypertrophy and ferroptosis. Proteomic analysis identified the upregulation of transcription factor high mobility group A2 (HMGA2) as a key mechanism in this ferroptotic process. Elevated HMGA2 levels exacerbated ferroptosis, evidenced by increased cell death, lipid peroxidation, ROS production, and reduced GPX4 expression. Furthermore, HMGA2 was shown to promote cardiomyocyte ferroptosis via binding to methyltransferase-like 14 (METTL14), which in turn enhanced ferroptosis in cardiomyocytes through solute carrier family 7 member 11 (SLC7A11) m6A modification. In vivo, a delivery system using neutrophil membrane (NM)-coated mesoporous silica nanoparticles (MSN) was developed to inhibit cardiac hypertrophy, underscoring the therapeutic potential of targeting ULK1. Overall, this study demonstrates that ULK1 promotes cardiac hypertrophy through HMGA2/METTL14/SLC7A11 axis-mediated cardiomyocyte ferroptosis, suggesting a novel therapeutic approach for cardiac hypertrophy.
Collapse
Affiliation(s)
- Meitian Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yuetong Sha
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Jiaxin Wang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Hanping Qi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Pilong Shi
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yongsheng Liu
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Man Jiang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Lina Ba
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yuhang Liu
- Department of Physiology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Yonggang Cao
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China
| | - Qianhui Zhang
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China.
| | - Hongli Sun
- Department of Pharmacology, Harbin Medical University-Daqing, Daqing, 163319, China.
| |
Collapse
|
2
|
Kot A, Koszewska D, Ochman B, Świętochowska E. Clinical Potential of Misshapen/NIKs-Related Kinase (MINK) 1-A Many-Sided Element of Cell Physiology and Pathology. Curr Issues Mol Biol 2024; 46:13811-13845. [PMID: 39727954 PMCID: PMC11727420 DOI: 10.3390/cimb46120826] [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: 10/15/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/28/2024] Open
Abstract
Misshapen/NIKs-related kinase (MINK) 1 belongs to the mammalian germinal center kinase (GCK) family. It contains the N-terminal, conserved kinase domain, a coiled-coil region, a proline-rich region, and a GCK, C-terminal domain with the Citron-NIK-Homology (CNH) domain. The kinase is an essential component of cellular signaling pathways, which include Wnt signaling, JNK signaling, pathways engaging Ras proteins, the Hippo pathway, and STRIPAK complexes. It thus contributes to regulating the cell cycle, apoptosis, cytoskeleton organization, cell migration, embryogenesis, or tissue homeostasis. MINK1 plays an important role in immunological responses, inhibiting Th17 and Th1 cell differentiation and regulating NLRP3 inflammasome function. It may be considered a link between ROS and the immunological system, and a potential antiviral target for human enteroviruses. The kinase has been implicated in the pathogenesis of sepsis, rheumatoid arthritis, asthma, SLE, and more. It is also involved in tumorigenesis and drug resistance in cancer. Silencing MINK1 reduces cancer cell migration, suggesting potential for new therapeutic approaches. Targeting MINK1 could be a promising treatment strategy for patients insensitive to current chemotherapies, and could improve their prognosis. Moreover, MINK1 plays an important role in the nervous system and the cardiovascular system development and function. The modulation of MINK1 activity could influence the course of neurodegenerative diseases, including Alzheimer's disease. Further exploration of the activity of the kinase could also help in gaining more insight into factors involved in thrombosis or congenital heart disease. This review aims to summarize the current knowledge on MINK1, highlight its therapeutic and prognostic potential, and encourage more studies in this area.
Collapse
Affiliation(s)
| | | | | | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland; (A.K.); (D.K.); (B.O.)
| |
Collapse
|
3
|
Brundage J, Barrios JP, Tison GH, Pirruccello JP. Genetics of Cardiac Aging Implicate Organ-Specific Variation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.02.24310874. [PMID: 39148824 PMCID: PMC11326326 DOI: 10.1101/2024.08.02.24310874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Heart structure and function change with age, and the notion that the heart may age faster for some individuals than for others has driven interest in estimating cardiac age acceleration. However, current approaches have limited feature richness (heart measurements; radiomics) or capture extraneous data and therefore lack cardiac specificity (deep learning [DL] on unmasked chest MRI). These technical limitations have been a barrier to efforts to understand genetic contributions to age acceleration. We hypothesized that a video-based DL model provided with heart-masked MRI data would capture a rich yet cardiac-specific representation of cardiac aging. In 61,691 UK Biobank participants, we excluded noncardiac pixels from cardiac MRI and trained a video-based DL model to predict age from one cardiac cycle in the 4-chamber view. We then computed cardiac age acceleration as the bias-corrected prediction of heart age minus the calendar age. Predicted heart age explained 71.1% of variance in calendar age, with a mean absolute error of 3.3 years. Cardiac age acceleration was linked to unfavorable cardiac geometry and systolic and diastolic dysfunction. We also observed links between cardiac age acceleration and diet, decreased physical activity, increased alcohol and tobacco use, and altered levels of 239 serum proteins, as well as adverse brain MRI characteristics. We found cardiac age acceleration to be heritable (h2g 26.6%); a genome-wide association study identified 8 loci related to linked to cardiomyopathy (near TTN, TNS1, LSM3, PALLD, DSP, PLEC, ANKRD1 and MYO18B) and an additional 16 loci (near MECOM, NPR3, KLHL3, HDGFL1, CDKN1A, ELN, SLC25A37, PI15, AP3M1, HMGA2, ADPRHL1, PGAP3, WNT9B, UHRF1 and DOK5). Of the discovered loci, 21 were not previously associated with cardiac age acceleration. Mendelian randomization revealed that lower genetically mediated levels of 6 circulating proteins (MSRA most strongly), as well as greater levels of 5 proteins (LXN most strongly) were associated with cardiac age acceleration, as were greater blood pressure and Lp(a). A polygenic score for cardiac age acceleration predicted earlier onset of arrhythmia, heart failure, myocardial infarction, and mortality. These findings provide a thematic understanding of cardiac age acceleration and suggest that heart- and vascular-specific factors are key to cardiac age acceleration, predominating over a more global aging program.
Collapse
Affiliation(s)
- James Brundage
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
| | - Joshua P. Barrios
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
| | - Geoffrey H. Tison
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Center for Biosignal Research, University of California San Francisco, San Francisco, CA, USA
| | - James P. Pirruccello
- Division of Cardiology, University of California San Francisco, San Francisco, CA, USA
- Bakar Computational Health Sciences Institute, University of California San Francisco, San Francisco, CA, USA
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Genetics Center, University of California San Francisco, San Francisco, CA, USA
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
- Center for Biosignal Research, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
4
|
Li F, Zhao X, Zhang Y, Zhuang Q, Wang S, Fang X, Xu T, Li X, Chen G. Exosomal circFAM63Bsuppresses bone regeneration of postmenopausal osteoporosis via regulating miR-578/HMGA2 axis. J Orthop Res 2024; 42:1244-1253. [PMID: 38151824 DOI: 10.1002/jor.25776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/23/2023] [Accepted: 12/24/2023] [Indexed: 12/29/2023]
Abstract
Postmenopausal osteoporosis (PMOP) affects hundreds of millions of elderly women worldwide. The imbalance between osteoblast-mediated bone formation and osteoclast-mediated bone resorption is the key factor in the progression of PMOP. Recently, exosomal circular RNAs have been considered as critical regulators in physiological and pathological progress. However, their roles in PMOP still require further exploration. Herein, we identified that the expression of exosomal circFAM63B significantly increased in PMOP patients and is closely related to bone density. We further demonstrated that circFAM63B inhibits osteogenic differentiation of bone marrow stromal cells and bone formation in ovariectomy mice by using a combination of in vitro and in vivo experiment strategies. Mechanistically, circFAM63B promotes HMGA2 expression by inhibiting miR-578, thereby suppressing bone repair. Our study proved that exosomal circFAM63B suppresses the bone regeneration of PMOP by regulating the miR-578/HMGA2 axis, which may provide new insights into the pathogenesis and development of PMOP. Knocking down exosomal circFAM63B could be regarded as a new strategy for the treatment of PMOP.
Collapse
Affiliation(s)
- Feng Li
- Department of Spine Surgery, the First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, China
| | - Xiaodong Zhao
- Department of Spine Surgery, the First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, China
| | - Yang Zhang
- Department of Spine Surgery, the First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, China
| | - Qingshan Zhuang
- Department of Spine Surgery, the First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, China
| | - Song Wang
- Department of Spine Surgery, the First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, China
| | - Xichi Fang
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopedic Surgery, Division of Hand& Foot and Microvascular Surgery, the First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Tao Xu
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopedic Surgery, Division of Hand& Foot and Microvascular Surgery, the First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| | - Xiaopeng Li
- Department of Spine Surgery, the First Affiliated Hospital of Weifang Medical University (Weifang People's Hospital), Weifang, China
- Clinical College of Orthopedics, Tianjin Medical University, Tianjin, China
| | - Gaoyang Chen
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopedic Surgery, Division of Hand& Foot and Microvascular Surgery, the First Affiliated Hospital of Southern University of Science and Technology (Shenzhen People's Hospital), Shenzhen, China
| |
Collapse
|
5
|
Wu CS, Chien YC, Yen CJ, Wu JY, Bai LY, Yu YL. EZH2-mediated epigenetic silencing of tumor-suppressive let-7c/miR-99a cluster by hepatitis B virus X antigen enhances hepatocellular carcinoma progression and metastasis. Cancer Cell Int 2023; 23:199. [PMID: 37689710 PMCID: PMC10493019 DOI: 10.1186/s12935-023-03002-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/25/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Hepatitis B virus (HBV)-encoded X antigen, HBx, assists in the development of hepatocellular carcinoma (HCC) through complex mechanisms. Our results provide new insights into the EZH2 epigenetic repression of let-7c that promotes HCC migration induced by HBx. Thus, let-7c and HMGA2 represent key diagnostic markers and potential therapeutic targets for the treatment of HBV-related HCC. RESULTS We investigated the epigenetic regulation of let-7c, an important representative miRNA in liver tumor metastasis, in human HCC cells to verify the effect of HBx. Based on quantitative PCR (qPCR) of mRNA isolated from tumor and adjacent non-tumor liver tissues of 24 patients with HBV-related HCC, EZH2 expression was significantly overexpressed in most HCC tissues (87.5%). We executed a miRNA microarray analysis in paired HBV-related HCC tumor and adjacent non-tumorous liver tissue from six of these patients and identified let-7c, miR-199a-3p, and miR-99a as being downregulated in the tumor tissue. Real-time PCR analysis verified significant downregulation of let-7c and miR-99a in both HepG2X and Hep3BX cells, which stably overexpress HBx, relative to parental cells. HBX enhanced EZH2 expression and attenuated let-7c expression to induce HMGA2 expression in the HCC cells. Knockdown of HMGA2 significantly downregulated the metastatic potential of HCC cells induced by HBx. CONCLUSIONS The deregulation of let-7c expression by HBx may indicate a potential novel pathway through deregulating cell metastasis and imply that HMGA2 might be used as a new prognostic marker and/or as an effective therapeutic target for HCC.
Collapse
Affiliation(s)
- Chen-Shiou Wu
- Institute of Translational Medicine and New Drug Development, Taichung, 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
| | - Yi-Chung Chien
- Institute of Translational Medicine and New Drug Development, Taichung, 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Chia-Jui Yen
- Division of Hematology and Oncology, Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, 70403, Taiwan
| | - Jia-Yan Wu
- Institute of Translational Medicine and New Drug Development, Taichung, 40402, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 40402, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan
| | - Li-Yuan Bai
- Division of Hematology and Oncology, China Medical University Hospital, Taichung, 40402, Taiwan.
| | - Yung-Luen Yu
- Institute of Translational Medicine and New Drug Development, Taichung, 40402, Taiwan.
- Center for Molecular Medicine, China Medical University Hospital, Taichung, 40402, Taiwan.
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 40402, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, 41354, Taiwan.
| |
Collapse
|
6
|
Sun X, Jiang Y, Li Q, Tan Q, Dong M, Cai B, Zhang D, Zhao Q. Quantitative proteomics analysis revealed the potential role of lncRNA Ftx in cardiomyocytes. Proteome Sci 2023; 21:2. [PMID: 36604692 PMCID: PMC9814437 DOI: 10.1186/s12953-022-00201-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 11/30/2022] [Indexed: 01/06/2023] Open
Abstract
OBJECTIVE This study aims to decode the proteomic signature of cardiomyocytes in response to lncRNA Ftx knockdown and overexpression via proteomic analysis, and to study the biological role of lncRNA Ftx in cardiomyocytes. METHODS: The expression level of the lncRNA Ftx in cardiomyocytes cultured in vitro was intervened, and the changes in protein levels in cardiomyocytes were quantitatively detected by liquid chromatography-mass spectrometry. The key molecules and pathways of the lncRNA-Ftx response were further examined by GO, KEGG, and protein interaction analysis. RESULTS A total of 2828 proteins are quantified. With a 1.5-fold change threshold, 32 upregulated proteins and 49 downregulated proteins are identified in the lncRNA Ftx overexpression group, while 67 up-regulated proteins and 54 down-regulated proteins are identified in the lncRNA Ftx knockdown group. Functional clustering analysis of differential genes revealed that the lncRNA Ftx is involved in regulating cardiomyocyte apoptosis and ferroptosis and improving cellular energy metabolism. In addition, Hub genes such as ITGB1, HMGA2, STAT3, GSS, and LPCAT3 are regulated downstream by lncRNA Ftx. CONCLUSION This study demonstrates that lncRNA Ftx plays a vital role in cardiomyocytes and may be involved in the occurrence and development of various myocardial diseases. It provides a potential target for clinical protection of the myocardium and reversal of myocardial fibrosis.
Collapse
Affiliation(s)
- Xiangfei Sun
- grid.460018.b0000 0004 1769 9639Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 9677 Jingshi Road, Jinan, 250021 Shandong China ,grid.27255.370000 0004 1761 1174Department of Cardiovascular Surgery, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021 Shandong China
| | - Ying Jiang
- grid.460018.b0000 0004 1769 9639Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, Jinan, 250021 Shandong China ,grid.27255.370000 0004 1761 1174Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Shandong 250021 Jinan, China
| | - Qingbao Li
- grid.460018.b0000 0004 1769 9639Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 9677 Jingshi Road, Jinan, 250021 Shandong China
| | - Qi Tan
- grid.460018.b0000 0004 1769 9639Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 9677 Jingshi Road, Jinan, 250021 Shandong China
| | - Mingliang Dong
- grid.460018.b0000 0004 1769 9639Department of Cardiovascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 9677 Jingshi Road, Jinan, 250021 Shandong China
| | - Bi’e Cai
- grid.479672.9Health Management Department of Preventive Treatment Center, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, No. 42, Wenhua West Road, Jinan, 250021 Shandong China
| | - Di Zhang
- grid.460018.b0000 0004 1769 9639Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, Jinan, 250021 Shandong China ,grid.27255.370000 0004 1761 1174Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Shandong 250021 Jinan, China
| | - Qi Zhao
- grid.460018.b0000 0004 1769 9639Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324 Jingwu Road, Jinan, 250021 Shandong China ,grid.27255.370000 0004 1761 1174Department of Gastroenterology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Shandong 250021 Jinan, China
| |
Collapse
|
7
|
Davidsen N, Ramhøj L, Kugathas I, Evrard B, Darde TA, Chalmel F, Svingen T, Rosenmai AK. PFOS disrupts key developmental pathways during hiPSC-derived cardiomyocyte differentiation in vitro. Toxicol In Vitro 2022; 85:105475. [PMID: 36116746 DOI: 10.1016/j.tiv.2022.105475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/17/2022] [Accepted: 09/12/2022] [Indexed: 11/26/2022]
Abstract
Exposure to perfluorooctanesulfonic acid (PFOS) has been associated with congenital heart disease (CHD) and decreased birth weight. PFOS exposure can disrupt signaling pathways relevant for cardiac development in stem cell-derived cardiomyocyte assays, such as the PluriBeat assay, where spheroids of human induced pluripotent stem cells (hiPSCs) differentiate into contracting cardiomyocytes. Notably, cell line origin can also affect how the assay responds to chemical exposure. Herein, we examined the effect of PFOS on cardiomyocyte differentiation by transcriptomics profiling of two different hiPSC lines to see if they exhibit a common pattern of disruption. Two stages of differentiation were investigated: the cardiac progenitor stage and the cardiomyocyte stage. Many differentially expressed genes (DEGs) were observed between cell lines independent of exposure. However, 135 DEGs were identified as common between the two cell lines. Of these, 10 DEGs were associated with GO-terms related to the heart. PFOS exposure disrupted multiple signaling pathways relevant to cardiac development, including WNT, TGF, HH, and EGF. Of these pathways, genes related to the non-canonical WNTCa2+ signaling was particularly affected. PFOS thus has the capacity to disrupt pathways important for cardiac development and function.
Collapse
Affiliation(s)
- Nichlas Davidsen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby DK-2800, Denmark.
| | - Louise Ramhøj
- National Food Institute, Technical University of Denmark, Kgs. Lyngby DK-2800, Denmark
| | - Indusha Kugathas
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000 Rennes, France
| | - Bertrand Evrard
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000 Rennes, France
| | | | - Frédéric Chalmel
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé, Environnement et Travail), UMR_S 1085, F-35000 Rennes, France
| | - Terje Svingen
- National Food Institute, Technical University of Denmark, Kgs. Lyngby DK-2800, Denmark
| | | |
Collapse
|
8
|
The proteasome activator REGγ promotes diabetic endothelial impairment by inhibiting HMGA2-GLUT1 pathway. Transl Res 2022; 246:33-48. [PMID: 35367424 DOI: 10.1016/j.trsl.2022.03.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 11/22/2022]
Abstract
Diabetic vascular endothelial impairment is one of the main causes of death in patients with diabetes lacking adequately defined mechanisms or effective treatments. REGγ, the 11S proteasome activator known to promote the degradation of cellular proteins in a ubiquitin- and ATP-independent manner, emerges as a new regulator in the cardiovascular system. Here, we found that REGγ was upregulated in streptozocin (STZ)-induced diabetic mouse aortic endothelium in vivo and high glucose (HG)-treated vascular endothelial cells (ECs) in vitro. REGγ deficiency ameliorated endothelial impairment in STZ-induced diabetic mice by protecting against a decline in cellular glucose uptake and associated vascular ECs dysfunction by suppressing high mobility group AT-hook 2 (HMGA2) decay. Mechanically, REGγ interacted with and degraded the transcription factor HMGA2 directly, leading to decreased HMGA2 transcriptional activity, subsequently lowered expression of glucose transporter type 1 (GLUT1), and reduced cellular glucose uptake, vascular endothelial dysfunction, and impaired diabetic endothelium. Ablation of endogenous GLUT1 or HMGA2 or overexpressing exogenous HMGA2 in vascular ECs significantly blocked or reestablished the REGγ-dependent action on cellular glucose uptake and vascular endothelial functions of HG stimulation in vitro. Furthermore, exogenously introducing HMGA2 improved diabetic mice endothelial impairment features caused by REGγ in vivo, thereby substantiating a REGγ-HMGA2-GLUT1 pathway in diabetic endothelial impairment. Our findings indicate that modulating REGγ-proteasome activity may be a potential therapeutic approach for diabetic disorders with endothelial impairment.
Collapse
|
9
|
Nomura S, Komuro I. Precision medicine for heart failure based on molecular mechanisms: The 2019 ISHR Research Achievement Award Lecture. J Mol Cell Cardiol 2021; 152:29-39. [PMID: 33275937 DOI: 10.1016/j.yjmcc.2020.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 11/02/2020] [Accepted: 11/24/2020] [Indexed: 10/22/2022]
Abstract
Heart failure is a leading cause of death, and the number of patients with heart failure continues to increase worldwide. To realize precision medicine for heart failure, its underlying molecular mechanisms must be elucidated. In this review summarizing the "The Research Achievement Award Lecture" of the 2019 XXIII ISHR World Congress held in Beijing, China, we would like to introduce our approaches for investigating the molecular mechanisms of cardiac hypertrophy, development, and failure, as well as discuss future perspectives.
Collapse
Affiliation(s)
- Seitaro Nomura
- Department of Cardiovascular Medicine, The University of Tokyo, Japan
| | - Issei Komuro
- Department of Cardiovascular Medicine, The University of Tokyo, Japan.
| |
Collapse
|
10
|
Cai ZL, Liu C, Yao Q, Xie QW, Hu TT, Wu QQ, Tang QZ. The pro-migration and anti-apoptosis effects of HMGA2 in HUVECs stimulated by hypoxia. Cell Cycle 2020; 19:3534-3545. [PMID: 33315504 DOI: 10.1080/15384101.2020.1850970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
High-mobility group AT-hook2 (HMGA2), serving as an architectural transcription factor, participates in plenty of biological processes. Our study is aimed at illustrating the effect of HMGA2 on hypoxia-induced HUVEC injury and the underlying mechanism. To induce hypoxia-related cell injury, HUVECs were exposed to hypoxic condition for 12-24 h. Molecular expression was determined by Western blot analysis, real-time PCR and immunofluorescence staining. Cell migration was monitored by wound healing assay and Transwell chamber assay. Cell proliferation and apoptosis were measured by MTT assay kits and TUNEL staining. In this study, we discovered that HMGA2 was upregulated in hypoxia-induced HUVECs. Overexpression of HMGA2 promoted cell migration, decreased the apoptosis ratio in response to hypoxia stimulation, while HMGA2 knockdown inhibited cell migration and accelerated apoptosis in HUVECs under hypoxic condition. Mechanistically, we found that HMGA2 induced increased expression of HIF-1α,VEGF, eNOS and AKT. eNOS knockdown significantly reduced HMGA2-mediated pro-migration effects, and AKT knockdown strikingly counteracted HMGA2-mediated anti-apoptotic effect. Hence, our data indicated that HMGA2 promoted cell migration by regulating HIF-1α/VGEF/eNOS signaling and prevented cell apoptosis by activating HIF-1α/VGEF/AKT signaling in HUVECs.
Collapse
Affiliation(s)
- Zhu-Lan Cai
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Chen Liu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qing-We Xie
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Tong-Tong Hu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qing-Qing Wu
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University , Wuhan, RP China.,Hubei Key Laboratory of Metabolic and Chronic Diseases , Wuhan, RP China
| |
Collapse
|
11
|
Weng LC, Hall AW, Choi SH, Jurgens SJ, Haessler J, Bihlmeyer NA, Grarup N, Lin H, Teumer A, Li-Gao R, Yao J, Guo X, Brody JA, Müller-Nurasyid M, Schramm K, Verweij N, van den Berg ME, van Setten J, Isaacs A, Ramírez J, Warren HR, Padmanabhan S, Kors JA, de Boer RA, van der Meer P, Sinner MF, Waldenberger M, Psaty BM, Taylor KD, Völker U, Kanters JK, Li M, Alonso A, Perez MV, Vaartjes I, Bots ML, Huang PL, Heckbert SR, Lin HJ, Kornej J, Munroe PB, van Duijn CM, Asselbergs FW, Stricker BH, van der Harst P, Kääb S, Peters A, Sotoodehnia N, Rotter JI, Mook-Kanamori DO, Dörr M, Felix SB, Linneberg A, Hansen T, Arking DE, Kooperberg C, Benjamin EJ, Lunetta KL, Ellinor PT, Lubitz SA. Genetic Determinants of Electrocardiographic P-Wave Duration and Relation to Atrial Fibrillation. CIRCULATION. GENOMIC AND PRECISION MEDICINE 2020; 13:387-395. [PMID: 32822252 PMCID: PMC7578098 DOI: 10.1161/circgen.119.002874] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BACKGROUND The P-wave duration (PWD) is an electrocardiographic measurement that represents cardiac conduction in the atria. Shortened or prolonged PWD is associated with atrial fibrillation (AF). We used exome-chip data to examine the associations between common and rare variants with PWD. METHODS Fifteen studies comprising 64 440 individuals (56 943 European, 5681 African, 1186 Hispanic, 630 Asian) and ≈230 000 variants were used to examine associations with maximum PWD across the 12-lead ECG. Meta-analyses summarized association results for common variants; gene-based burden and sequence kernel association tests examined low-frequency variant-PWD associations. Additionally, we examined the associations between PWD loci and AF using previous AF genome-wide association studies. RESULTS We identified 21 common and low-frequency genetic loci (14 novel) associated with maximum PWD, including several AF loci (TTN, CAND2, SCN10A, PITX2, CAV1, SYNPO2L, SOX5, TBX5, MYH6, RPL3L). The top variants at known sarcomere genes (TTN, MYH6) were associated with longer PWD and increased AF risk. However, top variants at other loci (eg, PITX2 and SCN10A) were associated with longer PWD but lower AF risk. CONCLUSIONS Our results highlight multiple novel genetic loci associated with PWD, and underscore the shared mechanisms of atrial conduction and AF. Prolonged PWD may be an endophenotype for several different genetic mechanisms of AF.
Collapse
Affiliation(s)
- Lu-Chen Weng
- Cardiovascular Rsrch Ctr, MGH, Boston
- Cardiovascular Disease Initiative, The Broad Inst of MIT & Harvard, Cambridge, MA
| | - Amelia Weber Hall
- Cardiovascular Rsrch Ctr, MGH, Boston
- Cardiovascular Disease Initiative, The Broad Inst of MIT & Harvard, Cambridge, MA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, The Broad Inst of MIT & Harvard, Cambridge, MA
| | - Sean J. Jurgens
- Cardiovascular Disease Initiative, The Broad Inst of MIT & Harvard, Cambridge, MA
| | - Jeffrey Haessler
- Fred Hutchinson Cancer Rsrch Ctr, Division of Public Health Sciences, Seattle WA
| | - Nathan A. Bihlmeyer
- McKusick-Nathans Dept of Genetic Medicine, Johns Hopkins Univ School of Med, Baltimore, MD
| | - Niels Grarup
- Novo Nordisk Foundation Ctr for Basic Metabolic Rsrch, Faculty of Health & Med Sciences, Univ of Copenhagen, Copenhagen, Denmark
| | - Honghuang Lin
- Boston Univ & NHLBI’s Framingham Heart Study, Framingham
- Section of Computational Biomedicine, Dept of Med, Boston Univ School of Med, Boston, MA
| | - Alexander Teumer
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Greifswald
- Inst for Community Med, Univ Medicine Greifswald, Greifswald, Germany
| | - Ruifang Li-Gao
- Dept of Clinical Epidemiology, Leiden Univ Medical Ctr, the Netherlands
| | - Jie Yao
- The Inst for Translational Genomics & Population Sciences at Harbor-UCLA Medical Ctr, Torrance
| | - Xiuqing Guo
- The Inst for Translational Genomics & Population Sciences at Harbor-UCLA Medical Ctr, Torrance
- Dept of Pediatrics, David Geffen School of Med at UCLA, Los Angeles, CA
| | - Jennifer A. Brody
- Cardiovascular Health Rsrch Unit, Dept of Med, Dept of Epidemiology, Univ of Washington
| | - Martina Müller-Nurasyid
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich
- Dept of Internal Med I (Cardiology), Hospital of the Ludwig-Maximilians-Univ (LMU) Munich, Munich
- Inst of Genetic Epidemiology, Helmholtz Zentrum München - German Rsrch Ctr for Environmental Health, Neuherberg, Germany
| | - Katharina Schramm
- Chair of Genetic Epidemiology, IBE, Faculty of Medicine, LMU Munich
- Dept of Internal Med I (Cardiology), Hospital of the Ludwig-Maximilians-Univ (LMU) Munich, Munich
- Inst of Genetic Epidemiology, Helmholtz Zentrum München - German Rsrch Ctr for Environmental Health, Neuherberg, Germany
| | - Niek Verweij
- Genomics plc, Oxford, UK
- Dept of Cardiology, Univ of Groningen & Univ Medical Ctr, Groningen
| | - Marten E. van den Berg
- Dept of Epidemiology, Division of Heart & Lungs, Univ of Utrecht, Univ Medical Ctr Utrecht
| | - Jessica van Setten
- Dept of Cardiology, Division of Heart & Lungs, Univ of Utrecht, Univ Medical Ctr Utrecht
| | - Aaron Isaacs
- CARIM School for Cardiovascular Diseases, Maastricht Univ, Maastricht, the Netherlands
- Dept of Physiology, Maastricht Univ, Maastricht, the Netherlands
| | - Julia Ramírez
- Nat Inst for Health Rsrch, Barts Cardiovascular Biomedical Rsrch Ctr, Barts & The London School of Med & Dentistry, Queen Mary Univ of London, London
- William Harvey Rsrch Inst, Barts & The London School of Med & Dentistry, Queen Mary Univ of London, London
| | - Helen R. Warren
- Nat Inst for Health Rsrch, Barts Cardiovascular Biomedical Rsrch Ctr, Barts & The London School of Med & Dentistry, Queen Mary Univ of London, London
- William Harvey Rsrch Inst, Barts & The London School of Med & Dentistry, Queen Mary Univ of London, London
| | - Sandosh Padmanabhan
- Inst of Cardiovascular & Medical Sciences, College of Medical, Veterinary & Life Sciences, Univ of Glasgow, Glasgow, UK
| | - Jan A. Kors
- Dept of Med Informatics, Erasmus Univ Medical Ctr, Rotterdam, the Netherlands
| | | | | | - Moritz F. Sinner
- Dept of Internal Med I (Cardiology), Hospital of the Ludwig-Maximilians-Univ (LMU) Munich, Munich
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Munich Heart Alliance, Munich
| | - Melanie Waldenberger
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Munich Heart Alliance, Munich
- Inst of Epidemiology, Helmholtz Zentrum München - German Rsrch Ctr for Environmental Health, Neuherberg, Germany
- Rsrch unit of Molecular Epidemiology, Helmholtz Zentrum München - German Rsrch Ctr for Environmental Health, Neuherberg, Germany
| | - Bruce M. Psaty
- Cardiovascular Health Rsrch Unit, Depts of Med, Epidemiology & Health Services, Dept of Epidemiology, Univ of Washington
- Kaiser Permanente Washington Health Rsrch Inst, Seattle, WA
| | - Kent D. Taylor
- The Inst for Translational Genomics & Population Sciences at Harbor-UCLA Medical Ctr, Torrance
- Dept of Pediatrics, David Geffen School of Med at UCLA, Los Angeles, CA
| | - Uwe Völker
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Greifswald
- Interfaculty Inst for Genetics & Functional Genomics, Univ Medicine Greifswald, Greifswald, Germany
| | - Jørgen K. Kanters
- Lab of Experimental Cardiology, Faculty of Health & Med Sciences, Univ of Copenhagen, Copenhagen, Denmark
| | - Man Li
- Division of Nephrology & Hypertensions, Dept of Internal Med, Univ of Utah School of Med, Salt Lake City, UT
| | - Alvaro Alonso
- Dept of Epidemiology, Rollins School of Public Health, Emory Univ, Atlanta, GA
| | | | - Ilonca Vaartjes
- Julius Ctr for Health Sciences & Primary Care, Univ Medical Ctr Utrecht, Utrecht Univ, the Netherlands
| | - Michiel L. Bots
- Julius Ctr for Health Sciences & Primary Care, Univ Medical Ctr Utrecht, Utrecht Univ, the Netherlands
| | | | - Susan R. Heckbert
- Cardiovascular Health Rsrch Unit, Dept of Epidemiology, Univ of Washington
| | - Henry J. Lin
- The Inst for Translational Genomics & Population Sciences at Harbor-UCLA Medical Ctr, Torrance
- Dept of Pediatrics, David Geffen School of Med at UCLA, Los Angeles, CA
| | - Jelena Kornej
- Boston Univ & NHLBI’s Framingham Heart Study, Framingham
| | - Patricia B. Munroe
- Nat Inst for Health Rsrch, Barts Cardiovascular Biomedical Rsrch Ctr, Barts & The London School of Med & Dentistry, Queen Mary Univ of London, London
- William Harvey Rsrch Inst, Barts & The London School of Med & Dentistry, Queen Mary Univ of London, London
| | - Cornelia M. van Duijn
- Dept of Epidemiology, Erasmus Univ Medical Ctr, Rotterdam, the Netherlands
- Nuffield Dept of Population Health, Medical Sciences Division, St. Cross College, Oxford Univ, Oxford
| | - Folkert W. Asselbergs
- Dept of Cardiology, Division of Heart & Lungs, Univ of Utrecht, Univ Medical Ctr Utrecht
- Health Data Rsrch UK & Inst of Health Informatics, Faculty of Population Health Sciences, Univ College London, London, UK
- Inst of Cardiovascular Science, Faculty of Population Health Sciences, Univ College London, London, UK
| | - Bruno H. Stricker
- Dept of Internal Medicine, Division of Heart & Lungs, Univ of Utrecht, Univ Medical Ctr Utrecht
- Dept of Med Informatics, Erasmus MC, Medical Ctr Rotterdam, Division of Heart & Lungs, Univ of Utrecht, Univ Medical Ctr Utrecht
- Inspectorate of Health Care
| | - Pim van der Harst
- Dept of Cardiology, Univ of Groningen & Univ Medical Ctr, Groningen
- Durrer Ctr for Cardiogenetic Rsrch, ICIN-Netherlands Heart Inst, Utrecht, the Netherlands
- Dept of Genetics, Univ of Groningen & Univ Medical Ctr, Groningen
| | - Stefan Kääb
- Dept of Internal Med I (Cardiology), Hospital of the Ludwig-Maximilians-Univ (LMU) Munich, Munich
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Munich Heart Alliance, Munich
| | - Annette Peters
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Munich Heart Alliance, Munich
- Inst of Epidemiology, Helmholtz Zentrum München - German Rsrch Ctr for Environmental Health, Neuherberg, Germany
- German Ctr for Diabetes Rsrch, Neuherberg, Germany
| | - Nona Sotoodehnia
- Cardiovascular Health Rsrch Unit, Dept of Med, Dept of Epidemiology, Univ of Washington
| | - Jerome I. Rotter
- The Inst for Translational Genomics & Population Sciences at Harbor-UCLA Medical Ctr, Torrance
- Depts of Pediatrics & Human Genetics, David Geffen School of Med at UCLA, Los Angeles, CA
| | - Dennis O. Mook-Kanamori
- Dept of Clinical Epidemiology, Leiden Univ Medical Ctr, the Netherlands
- Dept of Public Health & Primary Care, Leiden Univ Medical Ctr, the Netherlands
| | - Marcus Dörr
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Greifswald
- Dept of Internal Med B, Univ Medicine Greifswald, Greifswald, Germany
| | - Stephan B. Felix
- DZHK (German Ctr for Cardiovascular Rsrch), partner site Greifswald
- Dept of Internal Med B, Univ Medicine Greifswald, Greifswald, Germany
| | - Allan Linneberg
- Ctr for Clinical Rsrch & Prevention, Bispebjerg & Frederiksberg Hospital, Copenhagen, Denamrk
- Dept of Clinical Med, Faculty of Health & Med Sciences, Univ of Copenhagen, Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Ctr for Basic Metabolic Rsrch, Faculty of Health & Med Sciences, Univ of Copenhagen, Copenhagen, Denmark
| | - Dan E. Arking
- McKusick-Nathans Dept of Genetic Medicine, Johns Hopkins Univ School of Med, Baltimore, MD
| | - Charles Kooperberg
- Fred Hutchinson Cancer Rsrch Ctr, Division of Public Health Sciences, Seattle WA
| | - Emelia J. Benjamin
- Boston Univ & NHLBI’s Framingham Heart Study, Framingham
- Dept of Epidemiology, Boston Univ School of Public Health, Boston, MA
- Dept of Med, Boston Univ School of Med, Boston, MA
| | - Kathryn L. Lunetta
- Boston Univ & NHLBI’s Framingham Heart Study, Framingham
- Dept of Biostatistics, Boston Univ School of Public Health, Boston, MA
| | - Patrick T. Ellinor
- Cardiovascular Rsrch Ctr, MGH, Boston
- Cardiovascular Disease Initiative, The Broad Inst of MIT & Harvard, Cambridge, MA
- Cardiac Arrhythmia Service, MGH, Boston
| | - Steven A. Lubitz
- Cardiovascular Rsrch Ctr, MGH, Boston
- Cardiovascular Disease Initiative, The Broad Inst of MIT & Harvard, Cambridge, MA
- Cardiac Arrhythmia Service, MGH, Boston
| |
Collapse
|
12
|
HMGA Genes and Proteins in Development and Evolution. Int J Mol Sci 2020; 21:ijms21020654. [PMID: 31963852 PMCID: PMC7013770 DOI: 10.3390/ijms21020654] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/16/2022] Open
Abstract
HMGA (high mobility group A) (HMGA1 and HMGA2) are small non-histone proteins that can bind DNA and modify chromatin state, thus modulating the accessibility of regulatory factors to the DNA and contributing to the overall panorama of gene expression tuning. In general, they are abundantly expressed during embryogenesis, but are downregulated in the adult differentiated tissues. In the present review, we summarize some aspects of their role during development, also dealing with relevant studies that have shed light on their functioning in cell biology and with emerging possible involvement of HMGA1 and HMGA2 in evolutionary biology.
Collapse
|
13
|
Liu J, Hua RX, Cheng Y, Zhu J, Zhang J, Cheng J, Zhou H, Xia H, Bian J, He J. HMGA2 Gene rs8756 A>C Polymorphism Reduces Neuroblastoma Risk in Chinese Children: A Four-Center Case-Control Study. Onco Targets Ther 2020; 13:465-472. [PMID: 32021290 PMCID: PMC6970238 DOI: 10.2147/ott.s229975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 01/06/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Neuroblastoma, mainly affecting children, is a lethal malignancy arising from the developing sympathetic nervous system. The genetic etiology of neuroblastoma remains mostly obscure. High mobility group AT-hook 2 (HMGA2), an oncogenic gene, is up-regulated in many tumors. Single nucleotide polymorphisms (SNPs) often modify cancer susceptibility. However, no studies are investigating the association between HMGA2 SNPs and neuroblastoma susceptibility. METHODS We conducted a four-center case-control study to evaluate the association between three HMGA2 polymorphisms (rs6581658 A>G, rs8756 A>C and rs968697 T>C) and neuroblastoma susceptibility in a Chinese population with 505 cases and 1070 controls. Logistic regression was performed to evaluate the strength of the association. RESULTS We found that the rs8756 AC/CC genotypes were associated with a reduced neuroblastoma risk when compared to rs8756 AA genotype [Adjusted odds ratio (OR)=0.74, 95% confidence interval (CI)=0.56-0.99, P=0.039]. Carriers with 3 protective genotypes have lower neuroblastoma susceptibility than those without or with 0-2 protective genotypes. The stratified analysis revealed that the protective effects of rs8756 AC/CC genotypes were more predominant among children of age > 18 months, males, and subgroups with the tumor in the mediastinum. Furthermore, haplotype analysis uncovered that haplotype ACC significantly reduced neuroblastoma risk. CONCLUSION Our study indicated HMGA2 rs8756 A>C polymorphism is significantly associated with decreased neuroblastoma risk.
Collapse
Affiliation(s)
- Jiabin Liu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510623, Guangdong, People’s Republic of China
| | - Rui-Xi Hua
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510623, Guangdong, People’s Republic of China
- Department of Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou510080, Guangdong, People’s Republic of China
| | - Yun Cheng
- Department of Gynecology, Nanjing First Hospital Affiliated to Nanjing Medical University, Nanjing210006, Jiangsu, People’s Republic of China
| | - Jinhong Zhu
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510623, Guangdong, People’s Republic of China
- Department of Clinical Laboratory, Biobank, Harbin Medical University Cancer Hospital, Harbin150040, Heilongjiang, People’s Republic of China
| | - Jiao Zhang
- Department of Pediatric Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou450052, Henan, People’s Republic of China
| | - Jiwen Cheng
- Department of Pediatric Surgery, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an710004, Shaanxi, People’s Republic of China
| | - Haixia Zhou
- Department of Hematology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou325027, Zhejiang, People’s Republic of China
| | - Huimin Xia
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510623, Guangdong, People’s Republic of China
| | - Jun Bian
- Department of General Surgery, Xi’an Children’s Hospital, Xi’an Jiaotong University Affiliated Children’s Hospital, Xi’an710003, Shaanxi, People’s Republic of China
| | - Jing He
- Department of Pediatric Surgery, Guangzhou Institute of Pediatrics, Guangdong Provincial Key Laboratory of Research in Structural Birth Defect Disease, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou510623, Guangdong, People’s Republic of China
| |
Collapse
|
14
|
Chen CY, Choong OK, Liu LW, Cheng YC, Li SC, Yen CYT, Wu MR, Chiang MH, Tsang TJ, Wu YW, Lin LC, Chen YL, Lin WC, Hacker TA, Kamp TJ, Hsieh PCH. MicroRNA let-7-TGFBR3 signalling regulates cardiomyocyte apoptosis after infarction. EBioMedicine 2019; 46:236-247. [PMID: 31401194 PMCID: PMC6712055 DOI: 10.1016/j.ebiom.2019.08.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 12/18/2022] Open
Abstract
Background Myocardial infarction (MI) is a life-threatening disease, often leading to heart failure. Defining therapeutic targets at an early time point is important to prevent heart failure. Methods MicroRNA screening was performed at early time points after MI using paired samples isolated from the infarcted and remote myocardium of pigs. We also examined the microRNA expression in plasma of MI patients and pigs. For mechanistic studies, AAV9-mediated microRNA knockdown and overexpression were administrated in mice undergoing MI. Findings MicroRNAs let-7a and let-7f were significantly downregulated in the infarct area within 24 h post-MI in pigs. We also observed a reduction of let-7a and let-7f in plasma of MI patients and pigs. Inhibition of let-7 exacerbated cardiomyocyte apoptosis, induced a cardiac hypertrophic phenotype, and resulted in worsened left ventricular ejection fraction. In contrast, ectopic let-7 overexpression significantly reduced those phenotypes and improved heart function. We then identified TGFBR3 as a target of let-7, and found that induction of Tgfbr3 in cardiomyocytes caused apoptosis, likely through p38 MAPK activation. Finally, we showed that the plasma TGFBR3 level was elevated after MI in plasma of MI patients and pigs. Interpretation Together, we conclude that the let-7-Tgfbr3-p38 MAPK signalling plays an important role in cardiomyocyte apoptosis after MI. Furthermore, microRNA let-7 and Tgfbr3 may serve as therapeutic targets and biomarkers for myocardial damage. Fund Ministry of Science and Technology, National Health Research Institutes, Academia Sinica Program for Translational Innovation of Biopharmaceutical Development-Technology Supporting Platform Axis, Thematic Research Program and the Summit Research Program, Taiwan.
Collapse
Affiliation(s)
- Chen-Yun Chen
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Oi Kuan Choong
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Li-Wei Liu
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Yu-Che Cheng
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Sung-Chou Li
- Genomics and Proteomics Core Laboratory, Department of Medical Research, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | | | - Menq-Rong Wu
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Ming-Hsien Chiang
- Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tien-Jui Tsang
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Yen-Wen Wu
- Cardiology Division of Cardiovascular Medical Center and Department of Nuclear Medicine, Far Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Lung-Chun Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yuh-Lien Chen
- Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Chang Lin
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan
| | - Timothy A Hacker
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States
| | - Timothy J Kamp
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, United States
| | - Patrick C H Hsieh
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan; Department of Medicine, University of Wisconsin-Madison, Madison, WI, United States; Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI, United States; Institute of Medical Genomics and Proteomics, National Taiwan University College of Medicine, Taipei, Taiwan; Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, National Taiwan University Hospital, Taipei 100, Taiwan.
| |
Collapse
|
15
|
Liao YW, Ho BC, Chen MH, Yu SL. Host relieves lnc-IRAK3-3-sequestered miR-891b to attenuate apoptosis in Enterovirus 71 infection. Cell Microbiol 2019; 21:e13043. [PMID: 31099182 DOI: 10.1111/cmi.13043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 04/22/2019] [Accepted: 05/10/2019] [Indexed: 12/11/2022]
Abstract
Enterovirus 71 (EV71) is an emerging life-threatening pathogen particularly in the Asia-Pacific region. Apoptosis is a major pathogenic feature in EV71 infection. However, which molecular mechanism participating in EV71-induced apoptosis is not completely understood. Long noncoding RNAs (lncRNAs), a newly discovered class of regulatory RNA molecules, govern a wide range of biological functions through multiple regulatory mechanisms. Whether lncRNAs involved in EV71-induced apoptosis was investigated in this study. We conducted an apoptosis-oriented approach by integrating lncRNA and mRNA profilings. lnc-IRAK3-3 is down-regulated in EV71 infection and plays an important role in EV71 infection-induced apoptosis. Compensation of lnc-IRAK3-3 in EV71 infection promoted cell apoptosis wherein GADD45β expression was increased and further triggered caspase3 and PARP cleavage. Using bioinformatics analysis and functional assays, lnc-IRAK3-3 could functionally sequester miR-891b and GADD45β 3'UTR whereas miR-891b showed the inhibitory activity on GADD45β expression. Taken together, lnc-IRAK3-3 has the ability capturing miR-891b to enforce GADD45β expression and eventually promotes apoptosis. On the contrary, host cells suppress lnc-IRAK3-3 to relieve lnc-IRAK3-3-sequestered miR-891b, restrain GADD45β, and attenuate apoptosis in EV71 infection that prevent host cells from severe damages. We discover a new molecular mechanism by which host cells counteract EV71-induced apoptosis through the lnc-IRAK3-3/miR-891b/GADD45β axis partially.
Collapse
Affiliation(s)
- Yu-Wen Liao
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Bing-Ching Ho
- Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Min-Hsuan Chen
- Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan
| | - Sung-Liang Yu
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Centers of Genomic and Precision Medicine, National Taiwan University, Taipei, Taiwan.,Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan.,Institute of Medical Device and Imaging, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Clinical Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
16
|
Bertke MM, Dubiak KM, Cronin L, Zeng E, Huber PW. A deficiency in SUMOylation activity disrupts multiple pathways leading to neural tube and heart defects in Xenopus embryos. BMC Genomics 2019; 20:386. [PMID: 31101013 PMCID: PMC6525467 DOI: 10.1186/s12864-019-5773-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 05/03/2019] [Indexed: 02/08/2023] Open
Abstract
Background Adenovirus protein, Gam1, triggers the proteolytic destruction of the E1 SUMO-activating enzyme. Microinjection of an empirically determined amount of Gam1 mRNA into one-cell Xenopus embryos can reduce SUMOylation activity to undetectable, but nonlethal, levels, enabling an examination of the role of this post-translational modification during early vertebrate development. Results We find that SUMOylation-deficient embryos consistently exhibit defects in neural tube and heart development. We have measured differences in gene expression between control and embryos injected with Gam1 mRNA at three developmental stages: early gastrula (immediately following the initiation of zygotic transcription), late gastrula (completion of the formation of the three primary germ layers), and early neurula (appearance of the neural plate). Although changes in gene expression are widespread and can be linked to many biological processes, three pathways, non-canonical Wnt/PCP, snail/twist, and Ets-1, are especially sensitive to the loss of SUMOylation activity and can largely account for the predominant phenotypes of Gam1 embryos. SUMOylation appears to generate different pools of a given transcription factor having different specificities with this post-translational modification involved in the regulation of more complex, as opposed to housekeeping, processes. Conclusions We have identified changes in gene expression that underlie the neural tube and heart phenotypes resulting from depressed SUMOylation activity. Notably, these developmental defects correspond to the two most frequently occurring congenital birth defects in humans, strongly suggesting that perturbation of SUMOylation, either globally or of a specific protein, may frequently be the origin of these pathologies. Electronic supplementary material The online version of this article (10.1186/s12864-019-5773-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Michelle M Bertke
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA.,Present Address: College of Computer, Mathematical, and Natural Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Kyle M Dubiak
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Laura Cronin
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana, 46556, USA
| | - Erliang Zeng
- Department of Computer Science and Engineering, University of Notre Dame, Notre Dame, Indiana, 46556, USA.,Present Address: Division of Biostatistics and Computational Biology, Iowa Institute for Oral Health Research, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Preventive & Community Dentistry, College of Dentistry, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Biostatistics, University of Iowa, Iowa City, IA, 52242, USA.,Present Address: Department of Biomedical Engineering, University of Iowa, Iowa City, IA, 52242, USA
| | - Paul W Huber
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, 46556, USA. .,Harper Cancer Research Institute, University of Notre Dame, Notre Dame, Indiana, USA. .,Center for Stem Cells and Regenerative Medicine, University of Notre Dame, Notre Dame, Indiana, 46556, USA.
| |
Collapse
|
17
|
Kuan II, Lee CC, Chen CH, Lu J, Kuo YS, Wu HC. The extracellular domain of epithelial cell adhesion molecule (EpCAM) enhances multipotency of mesenchymal stem cells through EGFR-LIN28-LET7 signaling. J Biol Chem 2019; 294:7769-7786. [PMID: 30926604 DOI: 10.1074/jbc.ra119.007386] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/19/2019] [Indexed: 11/06/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are widely considered to be an attractive cell source for regenerative therapies, but maintaining multipotency and self-renewal in cultured MSCs is especially challenging. Hence, the development and mechanistic description of strategies that help promote multipotency in MSCs will be vital to future clinical use. Here, using an array of techniques and approaches, including cell biology, RT-quantitative PCR, immunoblotting, immunofluorescence, flow cytometry, and ChIP assays, we show that the extracellular domain of epithelial cell adhesion molecule (EpCAM) (EpEX) significantly increases the levels of pluripotency factors through a signaling cascade that includes epidermal growth factor receptor (EGFR), signal transducer and activator of transcription 3 (STAT3), and Lin-28 homolog A (LIN28) and enhances the proliferation of human bone marrow MSCs. Moreover, we found that EpEX-induced LIN28 expression reduces the expression of the microRNA LET7 and up-regulates that of the transcription factor high-mobility group AT-hook 2 (HMGA2), which activates the transcription of pluripotency factors. Surprisingly, we found that EpEX treatment also enhances osteogenesis of MSCs under differentiation conditions, as evidenced by increases in osteogenic markers, including Runt-related transcription factor 2 (RUNX2). Taken together, our results indicate that EpEX stimulates EGFR signaling and thereby context-dependently controls MSC states and activities, promoting cell proliferation and multipotency under maintenance conditions and osteogenesis under differentiation conditions.
Collapse
Affiliation(s)
- I-I Kuan
- From the Institute of Cellular and Organismic Biology and
| | - Chi-Chiu Lee
- From the Institute of Cellular and Organismic Biology and
| | - Chien-Hsu Chen
- From the Institute of Cellular and Organismic Biology and
| | - Jean Lu
- Genomic Research Center, Academia Sinica, Taipei 115 and
| | - Yuan-Sung Kuo
- the Department of Surgery, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Han-Chung Wu
- From the Institute of Cellular and Organismic Biology and .,Genomic Research Center, Academia Sinica, Taipei 115 and
| |
Collapse
|
18
|
Wong LL, Saw EL, Lim JY, Zhou Y, Richards AM, Wang P. MicroRNA Let-7d-3p Contributes to Cardiac Protection via Targeting HMGA2. Int J Mol Sci 2019; 20:ijms20071522. [PMID: 30934671 PMCID: PMC6480063 DOI: 10.3390/ijms20071522] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 03/21/2019] [Accepted: 03/25/2019] [Indexed: 01/01/2023] Open
Abstract
We tested the hypothesis that Let-7d-3p contributes to cardiac cell protection during hypoxic challenge. Myoblast H9c2 cells and primary neonatal rat ventricular cardiomyocytes (NRVM) were transfected with five selected miRNA mimics. Both cell lines were subjected to 0.2% oxygen hypoxia. The protective effects of these miRNAs were determined by assessment of cell metabolic activity by CCK8 assay and measurement of lactate dehydrogenase (LDH) release as a marker of cell injury. Apoptosis and autophagy flux were assessed by Annexin V/7-AAD double staining and the ratio of LC3 II/I with Baf-A1 treatment, an autophagy flux inhibitor, respectively. Luciferase-reporter assay, RT-qPCR and Western blots were performed to identify the changes of relevant gene targets. Among five miRNA mimic transfections, Let-7d-3p increased CCK8 activity, and decreased LDH release in both H9c2 and NRVM during hypoxia. Apoptosis was significantly reduced in H9c2 cells transfected with Let-7d-3p mimic. Autophagy and autophagy flux were not affected. In silico, mRNAs of HMGA2, YY1, KLF9, KLF12, and MEX3C are predicted targets for Let-7d-3p. Luciferase-reporter assay confirmed that Let-7d-3p bound directly to the 3’-UTR region of HMGA2, MEX3C, and YY1, the down-regulations of these mRNAs were verified in both H9c2 and NRVM. The protein expression of HMGA2, but not others, was downregulated in H9c2 and NRVM. It is known that HMGA2 is a strong apoptosis trigger through the blocking of DNA repair. Thus, we speculate that the anti-apoptotic effects of Let-7d-3p mimic during hypoxia challenge are due to direct targeting of HMGA2.
Collapse
Affiliation(s)
- Lee Lee Wong
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Eng Leng Saw
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Jia Yuen Lim
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Yue Zhou
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Arthur Mark Richards
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8014, New Zealand.
| | - Peipei Wang
- Cardiovascular Research Institute, National University Heart Centre, Singapore 117599, Singapore.
- Department of Medicine, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| |
Collapse
|
19
|
MicroRNA-34a-5p suppresses tumorigenesis and progression of glioma and potentiates Temozolomide-induced cytotoxicity for glioma cells by targeting HMGA2. Eur J Pharmacol 2019; 852:42-50. [PMID: 30851271 DOI: 10.1016/j.ejphar.2019.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 02/27/2019] [Accepted: 03/06/2019] [Indexed: 02/08/2023]
Abstract
Glioma is a frequently diagnosed brain tumors and Temozolomide (TMZ) is a common chemotherapeutic drug for glioma. High mobility group AT-hook 2 (HMGA2) was reported to be linked with glioma pathogenesis and Temozolomide (TMZ)-induced cytotoxicity. Our present study aimed to further search for the upstream regulatory microRNAs (miRNAs) of HMGA2 in glioma. RT-qPCR assay was conducted to measure the expression of HMGA2 mRNA and microRNA-34a-5p (miR-34a-5p). HMGA2 protein expression was examined by western blot assay. Cell proliferative ability and cell viability was assessed by CCK-8 assay. Cell migratory and invasive capacities were estimated by Transwell migration and invasion assay. Bioinformatics analysis and luciferase reporter assay was conducted to investigate the potential interaction between miR-34a-5p and HMGA2. Mouse xenograft experiments were performed to further test the roles of TMZ, miR-34a-5p and HMGA2, alone or in combination, in glioma tumorigenesis in vivo. We found HMGA2 expression was notably upregulated in glioma tissues and cells, and associated with glioma grade and poor prognosis. HMGA2 knockdown or miR-34a-5p overexpression inhibited migration, invasion, proliferation and enhanced TMZ-induced cytotoxicity in glioma cells. Moreover, HMGA2 was a target of miR-34a-5p. And, miR-34a-5p expression was remarkably reduced in glioma tissues and cells. MiR-34a-5p exerted its function through targeting HMGA2 in glioma cells. HMGA2 knockdown or miR-34a-5p overexpression inhibited tumor growth and enhanced TMZ-mediated anti-tumor effect in glioma xenograft models. We concluded MiR-34a-5p suppressed tumorigenesis and progression of glioma and potentiated TMZ-induced cytotoxicity for glioma cells by targeting HMGA2, deepening our understanding on molecular basis of HMGA2 in glioma.
Collapse
|
20
|
The protective effect of high mobility group protein HMGA2 in pressure overload-induced cardiac remodeling. J Mol Cell Cardiol 2019; 128:160-178. [PMID: 30711544 DOI: 10.1016/j.yjmcc.2019.01.027] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 01/07/2023]
Abstract
High mobility group protein AT-hook 2 (HMGA2), an architectural transcription factor, has previously been reported to play an essential role in regulating the expression of many genes through architectural remodeling processes. However, the effects of HMGA2 on cardiovascular disease, especial cardiac remodeling, is unclear. This study was aimed at investigating the functional role of HMGA2 in pressure overload-induced cardiac remodeling. Mice that were subjected to aortic banding (AB) for 8 weeks developed myocardial hypertrophy and cardiac dysfunction, which were associated with altered expression of HMGA2. Cardiac-specific expression of the human HMGA2 gene in mice with an adeno-related virus 9 delivery system ameliorated cardiac remodeling and improve cardiac function in response to pressure overload by activating PPARγ/NRF2 signaling. Knockdown of HMGA2 by AAV9-shHMGA2 accelerated cardiac remodeling after 1 weeks of AB surgery. Additionally, knockdown of heart PPARγ largely abolished HMGA2 overexpression-mediated cardioprotection. HMGA2-mediated cardiomyocyte protection was largely abrogated by knocking down NRF2 and inhibiting PPARγ in cardiomyocytes. PPARγ activation was mediated by C/EBPβ, which directly interacted with HMGA2. Knocking down C/EBPβ offset the effects of HMGA2 on PPARγ activation and cardioprotection. These findings show that the overexpression of HMGA2 ameliorates the remodeling response to pressure overload, and they also imply that the upregulation of HMGA2 may become a treatment strategy in cardiac pathologies.
Collapse
|
21
|
Mendonça Gorgulho C, Murthy P, Liotta L, Espina V, Lotze MT. Different measures of HMGB1 location in cancer immunology. Methods Enzymol 2019; 629:195-217. [DOI: 10.1016/bs.mie.2019.10.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
22
|
Saxena S, Gupta A, Shukla V, Rani V. Functional annotation of differentially expressed fetal cardiac microRNA targets: implication for microRNA-based cardiovascular therapeutics. 3 Biotech 2018; 8:494. [PMID: 30498667 DOI: 10.1007/s13205-018-1520-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/17/2018] [Indexed: 01/23/2023] Open
Abstract
Gene expression pattern of a failing heart depicts remarkable similarity with developing fetal heart. Elucidating genetic as well as epigenetic mechanisms regulating the gene expression during cardiac development will improve our understanding of cardiovascular diseases. In the present study, we aimed to validate and characterize differentially expressed known microRNAs (miRNA) obtained from next generation sequencing data of two fetal cardiac developmental stages (days 4th and 14th) from chicken (G. gallus domesticus) using bioinformatic approaches. Potential mRNA targets of individual miRNA were identified and classified according to their biological, cellular, and molecular functions. Functional annotation of putative target genes was performed to predict their association with cardiovascular diseases. We identified a total of 19 differentially expressed miRNAs between 4th and 14th day sample from the data sets obtained by next generation sequencing. A total of nearly 1522 potential targets ranging from 15 to 270 for each miRNA were predicted out of which 1221 were unique, while 301 were overlapping. Gene ontology and KEGG analysis revealed that majority of these target genes regulate critical cellular and molecular processes including transcriptional regulation, protein transport, signal transduction, matrix remodeling, Ras signaling, MAPK signaling, and TGF-beta signaling pathways indicating the complex nature of microRNA-mediated gene regulation during cardiogenesis. We found a significant association between potential target genes and cardiovascular diseases validating a link between fetal cardiac miRNAs and regulation of cardiovascular disease-related genes. These important findings may lay a foundation for further understanding the regulatory mechanisms operative in gene re-programming in the failing heart.
Collapse
|
23
|
Shi X, Huang T, Wang J, Liang Y, Gu C, Xu Y, Sun J, Lu Y, Sun K, Chen S, Yu Y. Next-generation sequencing identifies novel genes with rare variants in total anomalous pulmonary venous connection. EBioMedicine 2018; 38:217-227. [PMID: 30448225 PMCID: PMC6306349 DOI: 10.1016/j.ebiom.2018.11.008] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 10/22/2018] [Accepted: 11/03/2018] [Indexed: 12/30/2022] Open
Abstract
Background Total anomalous pulmonary venous connection (TAPVC) is recognized as a rare congenital heart defect (CHD). With a high mortality rate of approximately 80%, the survival rate and outcomes of TAPVC patients are not satisfactory. However, the genetic aetiology and mechanism of TAPVC remain elusive. This study aimed to investigate the underlying genomic risks of TAPVC through next-generation sequencing (NGS). Methods Rare variants were identified through whole exome sequencing (WES) of 78 sporadic TAPVC cases and 100 healthy controls using Fisher's exact test and gene-based burden test. We then detected candidate gene expression patterns in cells, pulmonary vein tissues, and embryos. Finally, we validated these genes using target sequencing (TS) in another 100 TAPVC cases. Findings We identified 42 rare variants of 7 genes (CLTCL1, CST3, GXYLT1, HMGA2, SNAI1, VAV2, ZDHHC8) in TAPVC cases compared with controls. These genes were highly expressed in human umbilical vein endothelial cells (HUVECs), mouse pulmonary veins and human embryonic hearts. mRNA levels of these genes in human pulmonary vein samples were significantly different between cases and controls. Through network analysis and expression patterns in zebrafish embryos, we revealed that SNAI1, HMGA2 and VAV2 are the most important genes for TAPVC. Interpretation Our study identifies novel candidate genes potentially related to TAPVC and elucidates the possible molecular pathogenesis of this rare congenital birth defect. Furthermore, SNAI1, HMGA2 and VAV2 are novel TAPVC candidate genes that have not been reported previously in either humans or animals. Fund National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Xin Shi
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing Wang
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Yulai Liang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Chang Gu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tong Ji University School of Medicine, Shanghai 200433, China
| | - Yuejuan Xu
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Jing Sun
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Yanan Lu
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China
| | - Kun Sun
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Sun Chen
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| | - Yu Yu
- Department of Pediatric Cardiovascular, Xin Hua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200092, China.
| |
Collapse
|
24
|
Metzger J, Rau J, Naccache F, Bas Conn L, Lindgren G, Distl O. Genome data uncover four synergistic key regulators for extremely small body size in horses. BMC Genomics 2018; 19:492. [PMID: 29940849 PMCID: PMC6019228 DOI: 10.1186/s12864-018-4877-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 06/18/2018] [Indexed: 02/06/2023] Open
Abstract
Background Miniature size in horses represents an extreme reduction of withers height that originated after domestication. In some breeds, it is a highly desired trait representing a breed- or subtype-specific feature. The genomic changes that emerged due to strong-targeted selection towards this distinct type remain unclear. Results Comparisons of whole-genome sequencing data from two Miniature Shetland ponies and one standard-sized Shetland pony, performed to elucidate genetic determinants for miniature size, revealed four synergistic variants, limiting withers height to 34.25 in. (87 cm). Runs of homozygosity regions were detected spanning these four variants in both the Miniature Shetland ponies and the standard-sized Shetland pony. They were shown to be characteristic of the Shetland pony breed, resulting in a miniature type under specific genotypic combinations. These four genetic variants explained 72% of the size variation among Shetland ponies and related breeds. The length of the homozygous regions indicate that they arose over 1000 years ago. In addition, a copy number variant was identified in DIAPH3 harboring a loss exclusively in ponies and donkeys and thus representing a potential height-associated variant. Conclusion This study reveals main drivers for miniature size in horses identified in whole genome data and thus provides relevant candidate genes for extremely short stature in mammals. Electronic supplementary material The online version of this article (10.1186/s12864-018-4877-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Julia Metzger
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Janina Rau
- Unit of Reproductive Medicine of the Clinics, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Fanny Naccache
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, 30559, Hannover, Germany
| | - Laura Bas Conn
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, 75007, Uppsala, Sweden
| | - Gabriella Lindgren
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, 75007, Uppsala, Sweden
| | - Ottmar Distl
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, 30559, Hannover, Germany.
| |
Collapse
|
25
|
Giancotti V, Bergamin N, Cataldi P, Rizzi C. Epigenetic Contribution of High-Mobility Group A Proteins to Stem Cell Properties. Int J Cell Biol 2018; 2018:3698078. [PMID: 29853899 PMCID: PMC5941823 DOI: 10.1155/2018/3698078] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Revised: 03/01/2018] [Accepted: 03/18/2018] [Indexed: 02/07/2023] Open
Abstract
High-mobility group A (HMGA) proteins have been examined to understand their participation as structural epigenetic chromatin factors that confer stem-like properties to embryonic stem cells (ESCs), induced pluripotent stem cells (iPSCs), and cancer stem cells (CSCs). The function of HMGA was evaluated in conjunction with that of other epigenetic factors such as histones and microRNAs (miRs), taking into consideration the posttranscriptional modifications (PTMs) of histones (acetylation and methylation) and DNA methylation. HMGA proteins were coordinated or associated with histone and DNA modification and the expression of the factors related to pluripotency. CSCs showed remarkable differences compared with ESCs and iPSCs.
Collapse
Affiliation(s)
- Vincenzo Giancotti
- Department of Life Science, University of Trieste, Trieste, Italy
- Trieste Proteine Ricerche, Palmanova, Udine, Italy
| | - Natascha Bergamin
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Palmina Cataldi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| | - Claudio Rizzi
- Division of Pathology, Azienda Ospedaliero-Universitaria, Udine, Italy
| |
Collapse
|
26
|
Bozzo M, Macrì S, Calzia D, Sgarra R, Manfioletti G, Ramoino P, Lacalli T, Vignali R, Pestarino M, Candiani S. The HMGA gene family in chordates: evolutionary perspectives from amphioxus. Dev Genes Evol 2017; 227:201-211. [PMID: 28474175 DOI: 10.1007/s00427-017-0581-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2016] [Accepted: 04/24/2017] [Indexed: 02/07/2023]
Abstract
High mobility group A proteins of vertebrates, HMGA1 and 2, are chromatin architectural factors involved in development, cell differentiation, and neoplastic transformation. Here, we characterize an amphioxus HMGA gene ortholog and analyze its expression. As a basal chordate, amphioxus is well placed to provide insights into the evolution of the HMGA gene family, particularly in the transition from invertebrates to vertebrates. Our phylogenetic analysis supports the basal position of amphioxus, echinoderm, and hemichordate HMGA sequences to those of vertebrate HMGA1 and HMGA2. Consistent with this, the genomic landscape around amphioxus HMGA shares features with both. Whole mount in situ hybridization shows that amphioxus HMGA mRNA is detectable from neurula stage onwards in both nervous and non-nervous tissues. This correlates with protein expression monitored immunocytochemically using antibodies against human HMGA2 protein, revealing especially high levels of expression in cells of the lamellar body, the amphioxus homolog of the pineal, suggesting that the gene may have, among its many functions, an evolutionarily conserved role in photoreceptor differentiation.
Collapse
Affiliation(s)
- Matteo Bozzo
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita (DISTAV), Università di Genova, viale Benedetto XV 5, 16132, Genoa, Italy
| | - Simone Macrì
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, SS12 Abetone e Brennero 4, 56127, Pisa, Italy
- Institute of Biotechnology, University of Helsinki, Biocenter 3, Viikinkaari 1, PO box 65, FI-00014, Helsinki, Finland
| | - Daniela Calzia
- Laboratorio di Biochimica, Dipartimento di Farmacia (DIFAR), Università di Genova, viale Benedetto XV 3, 16132, Genoa, Italy
| | - Riccardo Sgarra
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127, Trieste, Italy
| | - Guidalberto Manfioletti
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127, Trieste, Italy
| | - Paola Ramoino
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita (DISTAV), Università di Genova, viale Benedetto XV 5, 16132, Genoa, Italy
| | - Thurston Lacalli
- Biology Department, University of Victoria, BC V8W-3N5, Victoria, Canada
| | - Robert Vignali
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, SS12 Abetone e Brennero 4, 56127, Pisa, Italy
| | - Mario Pestarino
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita (DISTAV), Università di Genova, viale Benedetto XV 5, 16132, Genoa, Italy
| | - Simona Candiani
- Dipartimento di Scienze della Terra, dell'Ambiente e della Vita (DISTAV), Università di Genova, viale Benedetto XV 5, 16132, Genoa, Italy.
| |
Collapse
|
27
|
Sun X, Han Q, Luo H, Pan X, Ji Y, Yang Y, Chen H, Wang F, Lai W, Guan X, Zhang Q, Tang Y, Chu J, Yu J, Shou W, Deng Y, Li X. Profiling analysis of long non-coding RNAs in early postnatal mouse hearts. Sci Rep 2017; 7:43485. [PMID: 28266538 PMCID: PMC5339910 DOI: 10.1038/srep43485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/24/2017] [Indexed: 01/02/2023] Open
Abstract
Mammalian cardiomyocytes undergo a critical hyperplastic-to-hypertrophic growth transition at early postnatal age, which is important in establishing normal physiological function of postnatal hearts. In the current study, we intended to explore the role of long non-coding (lnc) RNAs in this transitional stage. We analyzed lncRNA expression profiles in mouse hearts at postnatal day (P) 1, P7 and P28 via microarray. We identified 1,146 differentially expressed lncRNAs with more than 2.0-fold change when compared the expression profiles of P1 to P7, P1 to P28, and P7 to P28. The neighboring genes of these differentially expressed lncRNAs were mainly involved in DNA replication-associated biological processes. We were particularly interested in one novel cardiac-enriched lncRNA, ENSMUST00000117266, whose expression was dramatically down-regulated from P1 to P28 and was also sensitive to hypoxia, paraquat, and myocardial infarction. Knockdown ENSMUST00000117266 led to a significant increase of neonatal mouse cardiomyocytes in G0/G1 phase and reduction in G2/M phase, suggesting that ENSMUST00000117266 is involved in regulating cardiomyocyte proliferative activity and is likely associated with hyperplastic-to-hypertrophic growth transition. In conclusion, our data have identified a large group of lncRNAs presented in the early postnatal mouse heart. Some of these lncRNAs may have important functions in cardiac hyperplastic-to-hypertrophic growth transition.
Collapse
Affiliation(s)
- Xiongshan Sun
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qi Han
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hongqin Luo
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaodong Pan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yan Ji
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yao Yang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Hanying Chen
- Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fangjie Wang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Wenjing Lai
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiao Guan
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qi Zhang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Yuan Tang
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Jianhong Chu
- Suzhou Institute of Blood and Marrow Transplantation, Soochow University, Suzhou, China
| | - Jianhua Yu
- Division of Hematology, Department of Internal Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Weinian Shou
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China.,Riley Heart Research Center, Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Youcai Deng
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Xiaohui Li
- Institute of Materia Medica, College of Pharmacy, Third Military Medical University, Chongqing, China.,Center of Translational Medicine, College of Pharmacy, Third Military Medical University, Chongqing, China
| |
Collapse
|
28
|
DeLaughter DM, Bick AG, Wakimoto H, McKean D, Gorham JM, Kathiriya IS, Hinson JT, Homsy J, Gray J, Pu W, Bruneau BG, Seidman JG, Seidman CE. Single-Cell Resolution of Temporal Gene Expression during Heart Development. Dev Cell 2016; 39:480-490. [PMID: 27840107 PMCID: PMC5198784 DOI: 10.1016/j.devcel.2016.10.001] [Citation(s) in RCA: 317] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/30/2016] [Accepted: 09/30/2016] [Indexed: 12/29/2022]
Abstract
Activation of complex molecular programs in specific cell lineages governs mammalian heart development, from a primordial linear tube to a four-chamber organ. To characterize lineage-specific, spatiotemporal developmental programs, we performed single-cell RNA sequencing of >1,200 murine cells isolated at seven time points spanning embryonic day 9.5 (primordial heart tube) to postnatal day 21 (mature heart). Using unbiased transcriptional data, we classified cardiomyocytes, endothelial cells, and fibroblast-enriched cells, thus identifying markers for temporal and chamber-specific developmental programs. By harnessing these datasets, we defined developmental ages of human and mouse pluripotent stem-cell-derived cardiomyocytes and characterized lineage-specific maturation defects in hearts of mice with heterozygous mutations in Nkx2.5 that cause human heart malformations. This spatiotemporal transcriptome analysis of heart development reveals lineage-specific gene programs underlying normal cardiac development and congenital heart disease.
Collapse
Affiliation(s)
| | - Alexander G. Bick
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Hiroko Wakimoto
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - David McKean
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Joshua M. Gorham
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Irfan S. Kathiriya
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA
- Department of Anesthesia and Perioperative Care, University of California, San Francisco
| | - John T. Hinson
- Division of Cardiovascular Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jason Homsy
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Jesse Gray
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - William Pu
- Department of Cardiology, Harvard Medical School, Boston Children's Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Benoit G. Bruneau
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA
- Cardiovascular Research Institute and Department of Pediatrics, University of California, San Francisco
| | - J. G. Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, Massachusetts, USA
- Howard Hughes Medical Institute and Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
29
|
Siahmansouri H, Somi MH, Babaloo Z, Baradaran B, Jadidi-Niaragh F, Atyabi F, Mohammadi H, Ahmadi M, Yousefi M. Effects of HMGA2 siRNA and doxorubicin dual delivery by chitosan nanoparticles on cytotoxicity and gene expression of HT-29 colorectal cancer cell line. J Pharm Pharmacol 2016; 68:1119-30. [DOI: 10.1111/jphp.12593] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 05/29/2016] [Indexed: 12/19/2022]
Abstract
Abstract
Objective
Over-expressions of HMGA2, vimentin and MMP-9 and downregulation of E-cadherin occur on colorectal cancer cells followed by a reduction in let-7 as a regulatory factor. In this study, we first used carboxymethyl dextran (CMD)–chitosan nanoparticles (ChNPs) platform to encapsulate HMGA2 siRNA and doxorubicin (DOX), and then, we evaluated the efficacy of the simultaneous delivery of siRNA/drug on viability and gene expression of HT-29 cell lines.
Methods
ChNPs characteristics were determined by a dynamic light scattering and zeta sizer. Morphology of loaded ChNPs was assessed by scanning electron microscopy, and Fourier transform infrared spectroscopy was used to confirm the conjugation of ChNP/siRNA/DOX/CMD. Cell viability and relative mRNA expression were evaluated by MTT assay and real-time PCR, respectively.
Key finding
The prepared ChNPs had high efficiency for siRNA and drug encapsulation (78% and 75%) and were stable against serum and heparin. ChNP/siRNA/DOX/CMD was more effective to induce tumour cell death and also could significantly reduce the expressions of HMGA2, vimentin as well as MMP-9 and increase E-cadherin expression.
Conclusion
In conclusion, our results revealed that dual delivery of a key gene siRNA and appropriate anticancer drug have great impact on the treatment of colorectal cancer.
Collapse
Affiliation(s)
- Homayoon Siahmansouri
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Hossein Somi
- Liver and Gastrointestinal Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zohreh Babaloo
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Jadidi-Niaragh
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Mohammadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Majid Ahmadi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
30
|
Wu H, Liang Y, Shen L, Shen L. MicroRNA-204 modulates colorectal cancer cell sensitivity in response to 5-fluorouracil-based treatment by targeting high mobility group protein A2. Biol Open 2016; 5:563-70. [PMID: 27095441 PMCID: PMC4874347 DOI: 10.1242/bio.015008] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are a conserved class of ∼22 nucleotide RNAs that playing important roles in various biological processes including chemoresistance. Recently, many studies have revealed that miR-204 is significantly attenuated in colorectal cancer (CRC), suggesting that this miRNA may have a function in CRC. However, whether miR-204 modulates chemosensitivity to 5-fluorouracil (5-Fu) in colorectal cancer is still unclear. In our present study, we discuss this possibility and the potential mechanism exerting this effect. We identified high mobility group protein A2 (HMGA2) as a novel direct target of miR-204 and showed that miR-204 expression was decreased while HMGA2 expression was increased in CRC cell lines. Additionally, both MiR-204 overexpression and HMGA2 inhibition attenuated cell proliferation, whereas forced expression of HMGA2 partly restored the inhibitory effect of miR-204 on HCT116 and SW480 cells. Moreover, the miR-204/HMGA2 axis modulated the resistance of tumor cells to 5-Fu in HCT-116 and SW480 colon cancer cells via activation of the PI3K/AKT pathway. These results demonstrate that the miR-204/HMGA2 axis could play a vital role in the 5-Fu resistance of colon cancer cells. Taken together, our present study elucidated that miR-204 upregulated 5-Fu chemosensitivity via the downregulation of HMGA2 in colorectal cancer and provided significant insight into the mechanism of 5-Fu resistance in colorectal cancer patients. More importantly, our present study suggested that miR-204 has potential as a therapeutic strategy for 5-Fu-resistant colorectal cancer. Summary: miR-204 upregulates 5-Fu chemosensitivity via the downregulation of HMGA2 in colorectal cancer and provides significant insight into the mechanism of 5-Fu resistance in colorectal cancer patients.
Collapse
Affiliation(s)
- Haijun Wu
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha 410008, China
| | - Yu Liang
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha 410008, China
| | - Lin Shen
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha 410008, China
| | - Liangfang Shen
- Department of Oncology, Xiangya Hospital, Central South University, 87 Xiang Ya Road, Changsha 410008, China
| |
Collapse
|
31
|
Jun-Hao ET, Gupta RR, Shyh-Chang N. Lin28 and let-7 in the Metabolic Physiology of Aging. Trends Endocrinol Metab 2016; 27:132-141. [PMID: 26811207 DOI: 10.1016/j.tem.2015.12.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 12/15/2015] [Accepted: 12/23/2015] [Indexed: 02/07/2023]
Abstract
The Lin28/let-7 molecular switch has emerged as a central regulator of growth signaling pathways and metabolic enzymes. Initially discovered to regulate developmental timing in the nematode, the Lin28/let-7 pathway of RNA regulation has gained prominence for its role in mammalian stem cells, cancer cells, tissue development, and aging. By regulating RNAs, the pathway coordinates cellular growth and cellular metabolism to influence metabolic physiology. Here, we review this regulatory mechanism and its impact on cancers, which reactivate Lin28, cardiovascular diseases, which implicate let-7, human genome-wide association studies (GWAS) of growth, and metabolic diseases, which implicate the Lin28/let-7 pathway. We also highlight questions relating to Barker's Hypothesis and the potential actions of the Lin28/let-7 pathway on programming long-lasting epigenetic effects.
Collapse
Affiliation(s)
- Elwin Tan Jun-Hao
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Renuka Ravi Gupta
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore
| | - Ng Shyh-Chang
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore.
| |
Collapse
|
32
|
SCHWARM FRANKP, UHLE FLORIAN, SCHÄNZER ANNE, ACKER TILL, STEIN MARCO, REINGES MARCUSH, WEISCHER CORNELIA, WEIGAND MARCUSA, UHL EBERHARD, KOLODZIEJ MALGORZATAA. High-mobility group AT-hook protein 2 expression and its prognostic significance in MGMT methylated and unmethylated glioblastoma. Int J Oncol 2016; 48:1485-92. [DOI: 10.3892/ijo.2016.3397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Accepted: 12/29/2015] [Indexed: 11/05/2022] Open
|
33
|
Macrì S, Simula L, Pellarin I, Pegoraro S, Onorati M, Sgarra R, Manfioletti G, Vignali R. Hmga2 is required for neural crest cell specification in Xenopus laevis. Dev Biol 2016; 411:25-37. [PMID: 26806704 DOI: 10.1016/j.ydbio.2016.01.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 12/23/2015] [Accepted: 01/20/2016] [Indexed: 12/12/2022]
Abstract
HMGA proteins are small nuclear proteins that bind DNA by conserved AT-hook motifs, modify chromatin architecture and assist in gene expression. Two HMGAs (HMGA1 and HMGA2), encoded by distinct genes, exist in mammals and are highly expressed during embryogenesis or reactivated in tumour progression. We here addressed the in vivo role of Xenopus hmga2 in the neural crest cells (NCCs). We show that hmga2 is required for normal NCC specification and development. hmga2 knockdown leads to severe disruption of major skeletal derivatives of anterior NCCs. We show that, within the NCC genetic network, hmga2 acts downstream of msx1, and is required for msx1, pax3 and snail2 activities, thus participating at different levels of the network. Because of hmga2 early effects in NCC specification, the subsequent epithelial-mesenchymal transition (EMT) and migration of NCCs towards the branchial pouches are also compromised. Strictly paralleling results on embryos, interfering with Hmga2 in a breast cancer cell model for EMT leads to molecular effects largely consistent with those observed on NCCs. These data indicate that Hmga2 is recruited in key molecular events that are shared by both NCCs and tumour cells.
Collapse
Affiliation(s)
- Simone Macrì
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Luca Simula
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Ilenia Pellarin
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Silvia Pegoraro
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Marco Onorati
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy
| | - Riccardo Sgarra
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Guidalberto Manfioletti
- Dipartimento di Scienze della Vita, Università degli Studi di Trieste, via Giorgieri 5, 34127 Trieste, Italy
| | - Robert Vignali
- Dipartimento di Biologia, Unità di Biologia Cellulare e dello Sviluppo, Università degli Studi di Pisa, SS12 Abetone e Brennero 4, 56127 Pisa, Italy.
| |
Collapse
|
34
|
Abstract
The molecular signatures of epigenetic regulation and chromatin architectures are fundamental to genetically determined biological processes. Covalent and post-translational chemical modification of the chromatin template can sensitize the genome to changing environmental conditions to establish diverse functional states. Recent interest and research focus surrounds the direct connections between metabolism and chromatin dynamics, which now represents an important conceptual challenge to explain many aspects of metabolic dysfunction. Several components of the epigenetic machinery require intermediates of cellular metabolism for enzymatic function. Furthermore, changes to intracellular metabolism can alter the expression of specific histone methyltransferases and acetyltransferases conferring widespread variations in epigenetic modification patterns. Specific epigenetic influences of dietary glucose and lipid consumption, as well as undernutrition, are observed across numerous organs and pathways associated with metabolism. Studies have started to define the chromatin-dependent mechanisms underlying persistent and pathophysiological changes induced by altered metabolism. Importantly, numerous recent studies demonstrate that gene regulation underlying phenotypic determinants of adult metabolic health is influenced by maternal and early postnatal diet. These emerging concepts open new perspectives to combat the rising global epidemic of metabolic disorders.
Collapse
Affiliation(s)
- Samuel T. Keating
- From the Epigenetics in Human Health and Disease Laboratory (S.T.K., A.E.-O.) and Epigenomics Profiling Facility (S.T.K., A.E.-O.), Baker IDI Heart & Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Department of Pathology, The University of Melbourne, Victoria, Australia (A.E.-O.); and Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia (A.E.-O.)
| | - Assam El-Osta
- From the Epigenetics in Human Health and Disease Laboratory (S.T.K., A.E.-O.) and Epigenomics Profiling Facility (S.T.K., A.E.-O.), Baker IDI Heart & Diabetes Institute, The Alfred Medical Research and Education Precinct, Melbourne, Victoria, Australia; Department of Pathology, The University of Melbourne, Victoria, Australia (A.E.-O.); and Central Clinical School, Department of Medicine, Monash University, Melbourne, Victoria, Australia (A.E.-O.)
| |
Collapse
|
35
|
Patterson M, Gaeta X, Loo K, Edwards M, Smale S, Cinkornpumin J, Xie Y, Listgarten J, Azghadi S, Douglass SM, Pellegrini M, Lowry WE. let-7 miRNAs can act through notch to regulate human gliogenesis. Stem Cell Reports 2014; 3:758-73. [PMID: 25316189 PMCID: PMC4235151 DOI: 10.1016/j.stemcr.2014.08.015] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2014] [Revised: 08/27/2014] [Accepted: 08/28/2014] [Indexed: 12/13/2022] Open
Abstract
It is clear that neural differentiation from human pluripotent stem cells generates cells that are developmentally immature. Here, we show that the let-7 plays a functional role in the developmental decision making of human neural progenitors, controlling whether these cells make neurons or glia. Through gain- and loss-of-function studies on both tissue and pluripotent derived cells, our data show that let-7 specifically regulates decision making in this context by regulation of a key chromatin-associated protein, HMGA2. Furthermore, we provide evidence that the let-7/HMGA2 circuit acts on HES5, a NOTCH effector and well-established node that regulates fate decisions in the nervous system. These data link the let-7 circuit to NOTCH signaling and suggest that this interaction serves to regulate human developmental progression. let-7 miRNAs influence developmental maturity of neural progenitors let-7 miRNAs act through HMGA2 and NOTCH to regulate gliogenesis HMGA2 expression regulates access of NICD to HES5 promoter Induction of let-7 miRNAs can accelerate oligodendrogenesis
Collapse
Affiliation(s)
- M Patterson
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - X Gaeta
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - K Loo
- Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - M Edwards
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA
| | - S Smale
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA
| | - J Cinkornpumin
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - Y Xie
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - J Listgarten
- Microsoft Research, 1100 Glendon Avenue Suite PH1, Los Angeles, CA 90024, USA
| | - S Azghadi
- Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - S M Douglass
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - M Pellegrini
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA
| | - W E Lowry
- Eli and Edythe Broad Center for Regenerative Medicine, UCLA, Box 957357, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, UCLA, 621 Charles E. Young Drive East, Los Angeles, CA 90095, USA; Molecular Biology Institute, UCLA, 611 Charles E. Young Drive East, Los Angeles, CA 90095, USA.
| |
Collapse
|
36
|
Karbassi E, Vondriska TM. How the proteome packages the genome for cardiovascular development. Proteomics 2014; 14:2115-26. [PMID: 25074278 DOI: 10.1002/pmic.201400131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 06/24/2014] [Accepted: 07/28/2014] [Indexed: 11/09/2022]
Abstract
The devastating impact of congenital heart defects has made mechanisms of vertebrate heart and vascular development an active area of study. Because myocyte death is a common feature of acquired cardiovascular diseases and the adult heart does not regenerate, the need exists to understand whether features of the developing heart and vasculature-which are more plastic-can be exploited therapeutically in the disease setting. We know that a core network of transcription factors governs commitment to the cardiovascular lineage, and recent studies using genetic loss-of-function approaches and unbiased genomic studies have revealed the role for various chromatin modulatory events. We reason that chromatin structure itself is a causal feature that influences transcriptome complexity along a developmental continuum, and the purpose of this article is to highlight the areas in which 'omics technologies have the potential to reveal new principles of phenotypic plasticity in development. We review the major mechanisms of chromatin structural regulation, highlighting what is known about their actions to control cardiovascular differentiation. We discuss emergent mechanisms of regulation that have been identified on the basis of genomic and proteomic studies of cardiac nuclei and identify current challenges to an integrated understanding of chromatin structure and cardiovascular phenotype.
Collapse
Affiliation(s)
- Elaheh Karbassi
- Departments of Anesthesiology, Medicine and Physiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | | |
Collapse
|
37
|
Wei J, Li H, Wang S, Li T, Fan J, Liang X, Li J, Han Q, Zhu L, Fan L, Zhao RC. let-7 enhances osteogenesis and bone formation while repressing adipogenesis of human stromal/mesenchymal stem cells by regulating HMGA2. Stem Cells Dev 2014; 23:1452-63. [PMID: 24617339 DOI: 10.1089/scd.2013.0600] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bone and fat cells share a common progenitor, stromal/mesenchymal stem cells (MSCs), that can differentiate into osteoblasts or adipocytes. Osteogenesis and adipogenesis of MSCs maintain homeostasis under physiological conditions. The disruption of this homeostasis leads to bone-related metabolic diseases. For instance, reduction in bone formation, which is usually accompanied by an increase in bone marrow adipogenesis, occurs with aging, immobility, or osteoporosis. Thus, it is crucial to gain an understanding of how osteogenic and adipogenic lineages of MSCs are regulated. Here, we present evidence that let-7 is a positive regulator of bone development. Using gain- and loss-of-function approaches, we demonstrate that let-7 markedly promotes osteogenesis and suppresses adipogenesis of MSCs in vitro. Moreover, let-7 could promote ectopic bone formation of MSCs in vivo. Subsequent studies further demonstrated that let-7's effects are mediated through the repression of high-mobility group AT-hook 2 (HMGA2) expression. RNAi depletion of HMGA2 could also enhance osteogenesis and repress adipogenesis. Overall, we found a novel role of let-7/HMGA2 axis in regulating the balance of osteogenesis and adipogenesis of MSCs. Thus, let-7 can be used as a novel therapeutic target for disorders that are associated with bone loss and adipocyte accumulation.
Collapse
Affiliation(s)
- Jianfeng Wei
- 1 Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Center of Excellence in Tissue Engineering, School of Basic Medicine Peking Union Medical College , Beijing, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Probing into the biological processes influenced by ESC factor and oncoprotein HMGA2 using iPSCs. Stem Cell Rev Rep 2014; 9:514-22. [PMID: 22547345 DOI: 10.1007/s12015-012-9373-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Induced pluripotent stem cells (iPSCs) are rapidly evolving into an important research tool due to their close resemblance with pluripotent embryonic stem cells (ESCs). Of particular interest at this point are iPSC applications in disease modeling and drug discovery/testing. The high mobility group AT-hook 2 (HMGA2) protein is a nonhistone chromatin factor normally expressed in ESCs and during early developmental stages. Aberrant HMGA2 expression is associated, for example, with abnormal body stature, diabetes mellitus, heart development and uterine leiomyomas. Furthermore, the protein is re-expressed in many primary tumor cells and plays an important role in metastasis. Here we used iPSC formation in conjunction with exogenous human HMGA2 expression to gain insight into biological functions of HMGA2. Gene expression profiling and gene ontology analyses showed that anatomical development and cell adhesion/differentiation processes are strongly affected by HMGA2. This could help to uncover, at the molecular level, some of the known phenotypic consequences of aberrant HMGA2 expression. Furthermore, our data showed that expression of key diabetes susceptibility genes is influenced by HMGA2, which revealed an interesting link to the recently indentified Lin28/let-7 pathway regulating mammalian glucose metabolism. Contrary to a previous report, our results indicate that HMGA2 is not involved in the regulation of telomerase gene expression. Finally, our data support a model in which tight regulation of intracellular HMGA2 levels is important both to maintain a pluripotent ESC state and to induce differentiation into certain cell lineages during later developmental stages.
Collapse
|
39
|
Zhou J, Dong X, Zhou Q, Wang H, Qian Y, Tian W, Ma D, Li X. microRNA expression profiling of heart tissue during fetal development. Int J Mol Med 2014; 33:1250-60. [PMID: 24604530 DOI: 10.3892/ijmm.2014.1691] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 02/18/2014] [Indexed: 01/08/2023] Open
Abstract
microRNAs (miRNAs) are important both in early cardiogenesis and in the process of heart maturation. The aim of this study was to determine the stage-specific expression of miRNAs in human fetal heart in order to identify valuable targets for further study of heart defects. Affymetrix microarrays were used to obtain miRNA expression profiles from human fetal heart tissue at 5, 7, 9 and 23 weeks of gestation. To identify differentially expressed miRNAs at each time-point, linear regression analysis by the R limma algorithm was employed. Hierarchical clustering analysis was conducted with Cluster 3.0 software. Gene Ontology analysis was carried out for miRNAs from different clusters. Commonalities in miRNA families and genomic localization were identified, and the differential expression of selected miRNAs from different clusters was verified by quantitative polymerase chain reaction (qPCR). A total of 703 miRNAs were expressed in human fetal heart. Of these, 288 differentially expressed miRNAs represented 5 clusters with different expression trends. Several clustered miRNAs also shared classification within miRNA families or proximal genomic localization. qPCR confirmed the expression patterns of selected miRNAs. miRNAs within the 5 clusters were predicted to target genes vital for heart development and to be involved in cellular signaling pathways that affect heart structure formation and heart-associated cellular events. In conclusion, to the best of our knowledge, this is the first miRNA expression profiling study of human fetal heart tissue. The stage-specific expression of specific miRNAs suggests potential roles at distinct time-points during fetal heart development.
Collapse
Affiliation(s)
- Jizi Zhou
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Xinran Dong
- Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Qiongjie Zhou
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| | - Huijun Wang
- Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Yanyan Qian
- Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Weidong Tian
- Department of Biostatistics and Computational Biology, School of Life Sciences, Fudan University, Shanghai, P.R. China
| | - Duan Ma
- Key Laboratory of Molecular Medicine, Ministry of Education, Department of Biochemistry and Molecular Biology, Shanghai Medical College, Fudan University, Shanghai, P.R. China
| | - Xiaotian Li
- Obstetrics and Gynecology Hospital, Fudan University, Shanghai, P.R. China
| |
Collapse
|
40
|
Ozturk N, Singh I, Mehta A, Braun T, Barreto G. HMGA proteins as modulators of chromatin structure during transcriptional activation. Front Cell Dev Biol 2014; 2:5. [PMID: 25364713 PMCID: PMC4207033 DOI: 10.3389/fcell.2014.00005] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2013] [Accepted: 02/07/2014] [Indexed: 01/12/2023] Open
Abstract
High mobility group (HMG) proteins are the most abundant non-histone chromatin associated proteins. HMG proteins bind to DNA and nucleosome and alter the structure of chromatin locally and globally. Accessibility to DNA within chromatin is a central factor that affects DNA-dependent nuclear processes, such as transcription, replication, recombination, and repair. HMG proteins associate with different multi-protein complexes to regulate these processes by mediating accessibility to DNA. HMG proteins can be subdivided into three families: HMGA, HMGB, and HMGN. In this review, we will focus on recent advances in understanding the function of HMGA family members, specifically their role in gene transcription regulation during development and cancer.
Collapse
Affiliation(s)
- Nihan Ozturk
- LOEWE Research Group Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research Bad Nauheim, Germany
| | - Indrabahadur Singh
- LOEWE Research Group Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research Bad Nauheim, Germany
| | - Aditi Mehta
- LOEWE Research Group Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research Bad Nauheim, Germany
| | - Guillermo Barreto
- LOEWE Research Group Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research Bad Nauheim, Germany
| |
Collapse
|
41
|
Li WY, Song YL, Xiong CJ, Lu PQ, Xue LX, Yao CX, Wang WP, Zhang SF, Zhang SF, Wei QX, Zhang YY, Zhao JM, Zang MX. Insulin induces proliferation and cardiac differentiation of P19CL6 cells in a dose-dependent manner. Dev Growth Differ 2013; 55:676-86. [PMID: 24020834 DOI: 10.1111/dgd.12075] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 07/27/2013] [Accepted: 08/04/2013] [Indexed: 12/29/2022]
Abstract
Insulin is a peptide hormone produced by beta cells of the pancreas. The roles of insulin in energy metabolism have been well studied, with most of the attention focused on glucose utilization, but the roles of insulin in cell proliferation and differentiation remain unclear. In this study, we observed for the first time that 10 nmol/L insulin treatment induces cell proliferation and cardiac differentiation of P19CL6 cells, whereas 50 and 100 nmol/L insulin treatment induces P19CL6 cell apoptosis and blocks cardiac differentiation of P19CL6 cells. By using real-time polymerase chain reaction (PCR) and Western blotting analysis, we found that the mRNA levels of cyclin D1 and α myosin heavy chain (α-MHC) are induced upon 10 nmol/L insulin stimulation and inhibited upon 50/100 nmol/L insulin treatment, whereas the mRNA levels of BCL-2-antagonist of cell death (BAD) exists a reverse trend. The similar results were observed in P19CL6 cells expressing GATA-6 or peroxisome proliferator-activated receptor α (PPARα). Our results identified the downstream targets of insulin, cyclin D1, BAD, α-MHC, and GATA-4, elucidate a novel molecular mechanism of insulin in promoting cell proliferation and differentiation.
Collapse
Affiliation(s)
- Wen-Yan Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, 450001, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Macrì S, Sgarra R, Ros G, Maurizio E, Zammitti S, Milani O, Onorati M, Vignali R, Manfioletti G. Expression and functional characterization of Xhmg-at-hook genes in Xenopus laevis. PLoS One 2013; 8:e69866. [PMID: 23936116 PMCID: PMC3723657 DOI: 10.1371/journal.pone.0069866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Accepted: 06/12/2013] [Indexed: 01/12/2023] Open
Abstract
High Mobility Group A proteins (HMGA1 and HMGA2) are architectural nuclear factors involved in development, cell differentiation, and cancer formation and progression. Here we report the cloning, developmental expression and functional analysis of a new multi-AT-hook factor in Xenopus laevis (XHMG-AT-hook) that exists in three different isoforms. Xhmg-at-hook1 and 3 isoforms, but not isoform 2, are expressed throughout the entire development of Xenopus, both in the maternal and zygotic phase. Localized transcripts are present in the animal pole in the early maternal phase; during the zygotic phase, mRNA can be detected in the developing central nervous system (CNS), including the eye, and in the neural crest. We show evidence that XHMG-AT-hook proteins differ from typical HMGA proteins in terms of their properties in DNA binding and in protein/protein interaction. Finally, we provide evidence that they are involved in early CNS development and in neural crest differentiation.
Collapse
Affiliation(s)
- Simone Macrì
- Department of Biology, University of Pisa, Pisa, Italy
| | - Riccardo Sgarra
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Gloria Ros
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Elisa Maurizio
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | - Salvina Zammitti
- Department of Life Sciences, University of Trieste, Trieste, Italy
| | | | - Marco Onorati
- Department of Biology, University of Pisa, Pisa, Italy
| | - Robert Vignali
- Department of Biology, University of Pisa, Pisa, Italy
- * E-mail: (GM); (RV)
| | | |
Collapse
|
43
|
Modak R, Basha J, Bharathy N, Maity K, Mizar P, Bhat AV, Vasudevan M, Rao VK, Kok WK, Natesh N, Taneja R, Kundu TK. Probing p300/CBP associated factor (PCAF)-dependent pathways with a small molecule inhibitor. ACS Chem Biol 2013; 8:1311-23. [PMID: 23570531 DOI: 10.1021/cb4000597] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PCAF (KAT2B) belongs to the GNAT family of lysine acetyltransferases (KAT) and specifically acetylates the histone H3K9 residue and several nonhistone proteins. PCAF is also a transcriptional coactivator. Due to the lack of a PCAF KAT-specific small molecule inhibitor, the exclusive role of the acetyltransferase activity of PCAF is not well understood. Here, we report that a natural compound of the hydroxybenzoquinone class, embelin, specifically inhibits H3Lys9 acetylation in mice and inhibits recombinant PCAF-mediated acetylation with near complete specificity in vitro. Furthermore, using embelin, we have identified the gene networks that are regulated by PCAF during muscle differentiation, further highlighting the broader regulatory functions of PCAF in muscle differentiation in addition to the regulation via MyoD acetylation.
Collapse
Affiliation(s)
- Rahul Modak
- Transcription
and Disease Laboratory,
Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore,
India 560064
| | - Jeelan Basha
- Transcription
and Disease Laboratory,
Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore,
India 560064
| | - Narendra Bharathy
- Department
of Physiology, Yong
Loo Lin School of Medicine, Block MD9, 2 Medical Drive, National University of Singapore, Singapore 117597
| | - Koustav Maity
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore, India 560012
| | - Pushpak Mizar
- Transcription
and Disease Laboratory,
Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore,
India 560064
| | - Akshay V. Bhat
- Transcription
and Disease Laboratory,
Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore,
India 560064
| | - Madavan Vasudevan
- Bionivid Technology [P] Ltd, 401 - 4 AB Cross, 1st Main, Kasturi Nagar,
East of NGEF, Bangalore, India 560043
| | - Vinay Kumar Rao
- Department
of Physiology, Yong
Loo Lin School of Medicine, Block MD9, 2 Medical Drive, National University of Singapore, Singapore 117597
| | - Wai Kay Kok
- Department
of Physiology, Yong
Loo Lin School of Medicine, Block MD9, 2 Medical Drive, National University of Singapore, Singapore 117597
| | - Nagashayana Natesh
- Central Government Health Scheme
Dispensary Number 3, Basavanagudi, Bangalore, India
| | - Reshma Taneja
- Department
of Physiology, Yong
Loo Lin School of Medicine, Block MD9, 2 Medical Drive, National University of Singapore, Singapore 117597
| | - Tapas K. Kundu
- Transcription
and Disease Laboratory,
Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore,
India 560064
| |
Collapse
|
44
|
Luo Y, Li W, Liao H. HMGA2 induces epithelial-to-mesenchymal transition in human hepatocellular carcinoma cells. Oncol Lett 2013; 5:1353-1356. [PMID: 23599793 PMCID: PMC3629159 DOI: 10.3892/ol.2013.1193] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2012] [Accepted: 02/05/2013] [Indexed: 11/13/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) is an important event during tumorigenesis. The human high-mobility group A2 (HMGA2) is a chromatin-binding protein, which contains three AT-hook domains that enable its binding to the minor groove of DNA. HMGA2 organizes protein complexes on enhancers of various genes to regulate gene expression and cell differentiation. The HMGA2 protein has been reported to be overexpressed in many types of cancer. It is not known, however, whether HMGA2 regulates EMT in human hepatocellular carcinoma (HCC) cell lines, and the mechanism(s) have not been fully elucidated. In this study, the expression of HMGA2 in five HCC cell lines was examined. The levels of HMGA2 expression among the five HCC cell lines coincided with their invasiveness. The variation in HMGA2 expression significantly correlated with the expression of several putative EMT markers. In addition, assessment of the invasive potential, following transfection with HMGA2-siRNA, demonstrated that the rate of cell migration was significantly reduced, suggesting that HMGA2 may be an important contributor to the invasion of tumor cells and that expression levels of HMGA2 influence the metastatic behavior of HCC cells. To further confirm the conclusion and explore the molecular mechanism through which HMGA2 induces EMT, we found that HMGA2 upregulates the expression of Twist and Snail in HCC cell lines. In conclusion, this present study is the first to show that HMGA2 effectively regulates EMT to induce invasion and metastasis in HCC cells. The function of HMGA2 as an oncoprotein may be associated with several important molecules involved in invasion and metastasis of cancer cells.
Collapse
Affiliation(s)
- Yizhou Luo
- Department of Oncology and Hematology, Jun Xie Hospital, Nanjing, Jiangsu 210002
| | | | | |
Collapse
|
45
|
Petersen JL, Mickelson JR, Rendahl AK, Valberg SJ, Andersson LS, Axelsson J, Bailey E, Bannasch D, Binns MM, Borges AS, Brama P, da Câmara Machado A, Capomaccio S, Cappelli K, Cothran EG, Distl O, Fox-Clipsham L, Graves KT, Guérin G, Haase B, Hasegawa T, Hemmann K, Hill EW, Leeb T, Lindgren G, Lohi H, Lopes MS, McGivney BA, Mikko S, Orr N, Penedo MCT, Piercy RJ, Raekallio M, Rieder S, Røed KH, Swinburne J, Tozaki T, Vaudin M, Wade CM, McCue ME. Genome-wide analysis reveals selection for important traits in domestic horse breeds. PLoS Genet 2013; 9:e1003211. [PMID: 23349635 PMCID: PMC3547851 DOI: 10.1371/journal.pgen.1003211] [Citation(s) in RCA: 184] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 11/15/2012] [Indexed: 02/04/2023] Open
Abstract
Intense selective pressures applied over short evolutionary time have resulted in homogeneity within, but substantial variation among, horse breeds. Utilizing this population structure, 744 individuals from 33 breeds, and a 54,000 SNP genotyping array, breed-specific targets of selection were identified using an F(ST)-based statistic calculated in 500-kb windows across the genome. A 5.5-Mb region of ECA18, in which the myostatin (MSTN) gene was centered, contained the highest signature of selection in both the Paint and Quarter Horse. Gene sequencing and histological analysis of gluteal muscle biopsies showed a promoter variant and intronic SNP of MSTN were each significantly associated with higher Type 2B and lower Type 1 muscle fiber proportions in the Quarter Horse, demonstrating a functional consequence of selection at this locus. Signatures of selection on ECA23 in all gaited breeds in the sample led to the identification of a shared, 186-kb haplotype including two doublesex related mab transcription factor genes (DMRT2 and 3). The recent identification of a DMRT3 mutation within this haplotype, which appears necessary for the ability to perform alternative gaits, provides further evidence for selection at this locus. Finally, putative loci for the determination of size were identified in the draft breeds and the Miniature horse on ECA11, as well as when signatures of selection surrounding candidate genes at other loci were examined. This work provides further evidence of the importance of MSTN in racing breeds, provides strong evidence for selection upon gait and size, and illustrates the potential for population-based techniques to find genomic regions driving important phenotypes in the modern horse.
Collapse
Affiliation(s)
- Jessica L Petersen
- College of Veterinary Medicine, University of Minnesota, St. Paul, Minnesota, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Li Z, Gilbert JA, Zhang Y, Zhang M, Qiu Q, Ramanujan K, Shavlakadze T, Eash JK, Scaramozza A, Goddeeris MM, Kirsch DG, Campbell KP, Brack AS, Glass DJ. An HMGA2-IGF2BP2 axis regulates myoblast proliferation and myogenesis. Dev Cell 2012. [PMID: 23177649 DOI: 10.1016/j.devcel.2012.10.019] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
A group of genes that are highly and specifically expressed in proliferating skeletal myoblasts during myogenesis was identified. Expression of one of these genes, Hmga2, increases coincident with satellite cell activation, and later its expression significantly declines correlating with fusion of myoblasts into myotubes. Hmga2 knockout mice exhibit impaired muscle development and reduced myoblast proliferation, while overexpression of HMGA2 promotes myoblast growth. This perturbation in proliferation can be explained by the finding that HMGA2 directly regulates the RNA-binding protein IGF2BP2. Add-back of IGF2BP2 rescues the phenotype. IGF2BP2 in turn binds to and controls the translation of a set of mRNAs, including c-myc, Sp1, and Igf1r. These data demonstrate that the HMGA2-IGF2BP2 axis functions as a key regulator of satellite cell activation and therefore skeletal muscle development.
Collapse
Affiliation(s)
- Zhizhong Li
- Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lee CT, Zhang L, Mounajjed T, Wu TT. High mobility group AT-hook 2 is overexpressed in hepatoblastoma. Hum Pathol 2012; 44:802-10. [PMID: 23134771 DOI: 10.1016/j.humpath.2012.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Revised: 07/29/2012] [Accepted: 08/05/2012] [Indexed: 11/30/2022]
Abstract
Hepatoblastoma is the most frequent malignant hepatic tumor in children. Expression of high mobility group AT-hook 2, an architectural nuclear factor and marker for hepatic progenitor cells, has not been studied in detail in hepatocellular neoplasms. Immunohistochemical stains using antibodies against high mobility group AT-hook 2, β-catenin, glypican-3, p53, and Ki-67 were performed in 15 hepatoblastomas, 15 fibrolamellar hepatocellular carcinomas, 34 hepatocellular carcinomas (12, ≤30 years old; 22, >30 years old), and 22 hepatic adenomas. High mobility group AT-hook 2 was expressed in all 15 hepatoblastomas, including all fetal and embryonal components, significantly higher than in 41.7% (5/12) of hepatocellular carcinomas of 30 years or younger (P = .001) and in 9% (2/22) of hepatocellular carcinomas of older than 30 years (P < .001). Aberrant β-catenin expression was detected in 80% (12/15) of hepatoblastomas, 41.6% (5/12) of hepatocellular carcinomas of 30 years or younger, and 18.2% (4/22) of hepatocellular carcinomas of older than 30 years. All 15 fibrolamellar hepatocellular carcinomas and 22 hepatic adenomas were negative for high mobility group AT-hook 2 or β-catenin. High mobility group AT-hook 2 and β-catenin expression correlated positively (P = .017; τ = 0.522) in 34 hepatocellular carcinomas. β-Catenin and glypican-3 exhibited a distinct spatial distribution within hepatoblastomas; glypican-3 was more frequently expressed in fetal components (P = .007), whereas β-catenin tended to be more frequently expressed in embryonal components (P = .062). In conclusion, high mobility group AT-hook 2 is expressed in all hepatoblastomas and could be used as a sensitive marker in their diagnosis. High mobility group AT-hook 2 was also expressed in a subset of hepatocellular carcinomas in association with β-catenin expression; this might represent a subtype of hepatocellular carcinoma with hepatic progenitor cell differentiation.
Collapse
Affiliation(s)
- Chung-Ta Lee
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan, Republic of China
| | | | | | | |
Collapse
|
48
|
Zhu S, Deng S, Ma Q, Zhang T, Jia C, Zhuo D, Yang F, Wei J, Wang L, Dykxhoorn DM, Hare JM, Goldschmidt-Clermont PJ, Dong C. MicroRNA-10A* and MicroRNA-21 modulate endothelial progenitor cell senescence via suppressing high-mobility group A2. Circ Res 2012; 112:152-64. [PMID: 23072816 DOI: 10.1161/circresaha.112.280016] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE Endothelial progenitor cells (EPCs) contribute to the regeneration of endothelium. Aging-associated senescence results in reduced number and function of EPCs, potentially contributing to increased cardiac risk, reduced angiogenic capacity, and impaired cardiac repair effectiveness. The mechanisms underlying EPC senescence are unknown. Increasing evidence supports the role of microRNAs in regulating cellular senescence. OBJECTIVE We aimed to determine whether microRNAs regulated EPC senescence and, if so, what the underlying mechanisms are. METHODS AND RESULTS To map the microRNA/gene expression signatures of EPC senescence, we performed microRNA profiling and microarray analysis in lineage-negative bone marrow cells from young and aged wild-type and apolipoprotein E-deficient mice. We identified 2 microRNAs, microRNA-10A* (miR-10A*), and miR-21, and their common target gene Hmga2 as critical regulators for EPC senescence. Overexpression of miR-10A* and miR-21 in young EPCs suppressed Hmga2 expression, caused EPC senescence, as evidenced by senescence-associated β-galactosidase upregulation, decreased self-renewal potential, increased p16(Ink4a)/p19(Arf) expression, and resulted in impaired EPC angiogenesis in vitro and in vivo, resembling EPCs derived from aged mice. In contrast, suppression of miR-10A* and miR-21 in aged EPCs increased Hmga2 expression, rejuvenated EPCs, resulting in decreased senescence-associated β-galactosidase expression, increased self-renewal potential, decreased p16(Ink4a)/p19(Arf) expression, and improved EPC angiogenesis in vitro and in vivo. Importantly, these phenotypic changes were rescued by miRNA-resistant Hmga2 cDNA overexpression. CONCLUSIONS miR-10A* and miR-21 regulate EPC senescence via suppressing Hmga2 expression and modulation of microRNAs may represent a potential therapeutic intervention in improving EPC-mediated angiogenesis and vascular repair.
Collapse
Affiliation(s)
- Shoukang Zhu
- Department of Medicine, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Cao JM, Li SQ, Zhang HW, Shi DL. High mobility group B proteins regulate mesoderm formation and dorsoventral patterning during zebrafish and Xenopus early development. Mech Dev 2012; 129:263-74. [DOI: 10.1016/j.mod.2012.07.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Revised: 06/29/2012] [Accepted: 07/02/2012] [Indexed: 10/28/2022]
|
50
|
Zhao W, Ji X, Zhang F, Li L, Ma L. Embryonic stem cell markers. Molecules 2012; 17:6196-236. [PMID: 22634835 PMCID: PMC6268870 DOI: 10.3390/molecules17066196] [Citation(s) in RCA: 131] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2012] [Revised: 03/31/2012] [Accepted: 05/04/2012] [Indexed: 02/07/2023] Open
Abstract
Embryonic stem cell (ESC) markers are molecules specifically expressed in ES cells. Understanding of the functions of these markers is critical for characterization and elucidation for the mechanism of ESC pluripotent maintenance and self-renewal, therefore helping to accelerate the clinical application of ES cells. Unfortunately, different cell types can share single or sometimes multiple markers; thus the main obstacle in the clinical application of ESC is to purify ES cells from other types of cells, especially tumor cells. Currently, the marker-based flow cytometry (FCM) technique and magnetic cell sorting (MACS) are the most effective cell isolating methods, and a detailed maker list will help to initially identify, as well as isolate ESCs using these methods. In the current review, we discuss a wide range of cell surface and generic molecular markers that are indicative of the undifferentiated ESCs. Other types of molecules, such as lectins and peptides, which bind to ESC via affinity and specificity, are also summarized. In addition, we review several markers that overlap with tumor stem cells (TSCs), which suggest that uncertainty still exists regarding the benefits of using these markers alone or in various combinations when identifying and isolating cells.
Collapse
Affiliation(s)
- Wenxiu Zhao
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| | - Xiang Ji
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Fangfang Zhang
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Liang Li
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
- Department of Biological Sciences and Biotechnology, Tsinghua University, Beijing 100084, China
| | - Lan Ma
- Life Science Division, Graduate School at Shenzhen, Tsinghua University, Shenzhen 518055, China; (W.Z.); (X.J.); (F.Z.); (L.L.)
| |
Collapse
|