1
|
Ren L, Wang Y, Ren Y, Li G, Sang N. Phenanthrene perturbs hematopoietic development and causes hematopoietic defects in zebrafish. J Environ Sci (China) 2025; 151:573-581. [PMID: 39481963 DOI: 10.1016/j.jes.2024.02.018] [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: 08/29/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 11/03/2024]
Abstract
Phenanthrene (Phe) is one of the common polycyclic aromatic hydrocarbons in the environment, and recent studies show that it can cause cardiac developmental toxicity and immunotoxicity. However, it is still unknown whether it can affect the hematopoietic development in aquatic organisms. To address this question, zebrafish (Danio rerio) were chronically exposed to Phe at different concentrations. We found that Phe caused structural damage to the renal tubules in the kidney, induced malformed erythrocytes in peripheral blood, and decreased the proportion of myeloid cells in adult zebrafish, suggesting possible negative impacts that Phe posed to hematopoietic development. Then, using in situ hybridization technology, we found that Phe decreased the expression of primitive hematopoietic marker genes, specifically gata1 and pu.1, accompanied by an obstruction of primitive erythrocyte circulation. Furthermore, Phe impaired definitive hematopoiesis, increased aberrations of the transient hematopoietic site (PBI), and reduced the generation of hematopoietic stem cells, ultimately influencing the number of erythrocytes and myeloid cells. The findings suggested that Phe could induce hematopoietic toxicity in zebrafish embryos and pose unknown ecological risks.
Collapse
Affiliation(s)
- Lingyu Ren
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Yue Wang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Ying Ren
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| | - Guangke Li
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China.
| | - Nan Sang
- College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan 030006, China
| |
Collapse
|
2
|
Fahim SA, Ragheb M, Fayed IH, Osama A, Karam A, Magdeldin S, Metwale R, Elsayed MDAA, Abdellatif A, Sadek HA, El Sobky SA, El-Ekiaby N, Fawzy IO, Abdelaziz AI. Interaction Between Malat1 and miR-499-5p Regulates Meis1 Expression and Function with a Net Impact on Cell Proliferation. Cells 2025; 14:125. [PMID: 39851553 PMCID: PMC11764005 DOI: 10.3390/cells14020125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/15/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025] Open
Abstract
Meis1 is a transcription factor involved in numerous functions including development and proliferation and has been previously shown to harness cell cycle progression. In this study, we used in silico analysis to predict that miR-499-5p targets Meis1 and that Malat1 sponges miR-499-5p. For the first time, we demonstrated that the overexpression of miR-499-5p led to the downregulation of Meis1 mRNA and protein in C166 cells by directly binding to its 3'UTR. Moreover, knocking down Malat1 increased miR-499-5p expression, subsequently suppressing Meis1. Through BrdU incorporation assay, we showed that the knockdown of Malat1, Meis1, or mimicking with miR-499-5p promoted cell proliferation. Enrichment analyses on proteins identified via mass spectrometry after manipulating Malat1, miR-499-5p, or Meis1 revealed a multitude of differentially expressed proteins related to cell cycle, cell division, and key pathways like Wnt and mTOR, essential for cell proliferation. Collectively, our findings confirm that Malat1 sponges miR-499-5p, regulating Meis1, and that Malat1/miR-499-5p/Meis1 could potentially form an axis that has a pivotal influence on cellular proliferation.
Collapse
Affiliation(s)
- Salma A. Fahim
- School of Medicine, Newgiza University (NGU), Giza 12577, Egypt
- Biotechnology Program, American University in Cairo, Cairo 11835, Egypt
| | - Manon Ragheb
- School of Medicine, Newgiza University (NGU), Giza 12577, Egypt
- Biotechnology Program, American University in Cairo, Cairo 11835, Egypt
| | | | - Aya Osama
- Proteomics and Metabolomics Unit, Basic Research Department, Children’s Cancer Hospital 57357 Cairo, (CCHE-57357), Cairo 11562, Egypt
| | - Ahmed Karam
- Proteomics and Metabolomics Unit, Basic Research Department, Children’s Cancer Hospital 57357 Cairo, (CCHE-57357), Cairo 11562, Egypt
| | - Sameh Magdeldin
- Proteomics and Metabolomics Unit, Basic Research Department, Children’s Cancer Hospital 57357 Cairo, (CCHE-57357), Cairo 11562, Egypt
- Department of Physiology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| | - Rana Metwale
- School of Medicine, Newgiza University (NGU), Giza 12577, Egypt
| | - Mohamed Dief Allah Abdalmoneam Elsayed
- School of Medicine, Newgiza University (NGU), Giza 12577, Egypt
- Department of Anatomy and Embryology, Faculty of Medicine, Cairo University, Cairo 11562, Egypt
| | - Ahmed Abdellatif
- Biotechnology Program, American University in Cairo, Cairo 11835, Egypt
| | - Hesham A. Sadek
- Division of Cardiology, University of Arizona College of Medicine, Tucson, AR 85721, USA
- Division of Cardiology, Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 85004, USA
| | | | - Nada El-Ekiaby
- School of Medicine, Newgiza University (NGU), Giza 12577, Egypt
| | | | | |
Collapse
|
3
|
Liu XY, Chen B, Zhang R, Zhang MQ, Ma YY, Han Y, Jiang JD, Zhang JP. Atorvastatin-induced intracerebral hemorrhage is inhibited by berberine in zebrafish. J Appl Toxicol 2024; 44:1198-1213. [PMID: 38639436 DOI: 10.1002/jat.4614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/18/2024] [Accepted: 04/02/2024] [Indexed: 04/20/2024]
Abstract
Intracerebral hemorrhage (ICH), for which there are currently no effective preventive or treatment methods, has a very high fatality rate. Statins, such as atorvastatin (ATV), are the first-line drugs for regulating blood lipids and treating hyperlipidemia-related cardiovascular diseases. However, ATV-associated ICH has been reported, although its incidence is rare. In this study, we aimed to investigate the protective action and mechanisms of berberine (BBR) against ATV-induced brain hemorrhage. We established an ICH model in zebrafish induced by ATV (2 μM) and demonstrated the effects of BBR (10, 50, and 100 μM) on ICH via protecting the vascular network using hemocyte staining and three transgenic zebrafish. BBR was found to reduce brain inflammation and locomotion injury in ICH-zebrafish. Mechanism research showed that ATV increased the levels of VE-cadherin and occludin proteins but disturbed their localization at the cell membrane by abnormal phosphorylation, which decreased the number of intercellular junctions between vascular endothelial cells (VECs), disrupting the integrity of vascular walls. BBR reversed the effects of ATV by promoting autophagic degradation of phosphorylated VE-cadherin and occludin in ATV-induced VECs examined by co-immunoprecipitation (co-IP). These findings provide crucial insights into understanding the BBR mechanisms involved in the maintenance of vascular integrity and in mitigating adverse reactions to ATV.
Collapse
Affiliation(s)
- Xin-Yan Liu
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bo Chen
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao-Qing Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuan-Yuan Ma
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Han
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian-Dong Jiang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Pu Zhang
- Key Laboratory of Biotechnology of Antibiotics, the National Health Commission (NHC), Beijing Key Laboratory of Antimicrobial Agents, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Fuentevilla-Alvarez G, Soto ME, Robles-Herrera GJ, Vargas-Alarcón G, Sámano R, Meza-Toledo SE, Huesca-Gómez C, Gamboa R. Analysis of Circulating miRNA Expression Profiles in Type 2 Diabetes Patients with Diabetic Foot Complications. Int J Mol Sci 2024; 25:7078. [PMID: 39000190 PMCID: PMC11241130 DOI: 10.3390/ijms25137078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/25/2024] [Indexed: 07/16/2024] Open
Abstract
Type 2 diabetes mellitus (T2DM) is associated with various complications, including diabetic foot, which can lead to significant morbidity and mortality. Non-healing foot ulcers in diabetic patients are a major risk factor for infections and amputations. Despite conventional treatments, which have limited efficacy, there is a need for more effective therapies. MicroRNAs (miRs) are small non-coding RNAs that play a role in gene expression and have been implicated in diabetic wound healing. miR expression was analyzed through RT-qPCR in 41 diabetic foot Mexican patients and 50 controls. Diabetic foot patients showed significant increases in plasma levels of miR-17-5p (p = 0.001), miR-191-5p (p = 0.001), let-7e-5p (p = 0.001), and miR-33a-5p (p = 0.005) when compared to controls. Elevated levels of miR-17, miR-191, and miR-121 correlated with higher glucose levels in patients with diabetic foot ulcers (r = 0.30, p = 0.004; r = 0.25, p = 0.01; and r = 0.21, p = 0.05, respectively). Levels of miR-17 showed the highest diagnostic potential (AUC 0.903, p = 0.0001). These findings underscore the possible role of these miRs in developing diabetes complications. Our study suggests that high miR-17, miR-191, and miR-121 expression is strongly associated with higher glucose levels and the development of diabetic foot ulcers.
Collapse
Affiliation(s)
- Giovanny Fuentevilla-Alvarez
- Endocrinology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico;
| | - María Elena Soto
- Research Direction, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (M.E.S.); (G.V.-A.)
- Cardiovascular Line in American British Cowdary (ABC) Medical Center, Sur 136 No. 116 Col. Las Américas, Mexico City 01120, Mexico
| | - Gustavo Jaziel Robles-Herrera
- Phisiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (G.J.R.-H.); (C.H.-G.)
| | - Gilberto Vargas-Alarcón
- Research Direction, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (M.E.S.); (G.V.-A.)
| | - Reyna Sámano
- Coordinación de Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Mexico City 11000, Mexico;
| | - Sergio Enrique Meza-Toledo
- Biochemistry Department, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional (IPN), Mexico City 11340, Mexico;
| | - Claudia Huesca-Gómez
- Phisiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (G.J.R.-H.); (C.H.-G.)
| | - Ricardo Gamboa
- Phisiology Department, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No. 1. Col. Sección XVI, Mexico City 14080, Mexico; (G.J.R.-H.); (C.H.-G.)
| |
Collapse
|
5
|
Javanshir E, Ebrahimi ZJ, Mirzohreh ST, Ghaffari S, Banisefid E, Alamdari NM, Roshanravan N. Disparity of gene expression in coronary artery disease: insights from MEIS1, HIRA, and Myocardin. Mol Biol Rep 2024; 51:712. [PMID: 38824221 DOI: 10.1007/s11033-024-09657-5] [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: 03/31/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
INTRODUCTION Coronary artery disease (CAD) in young adults can have devastating consequences. The cardiac developmental gene MEIS1 plays important roles in vascular networks and heart development. This gene effects on the regeneration capacity of the heart. Considering role of MEIS1 in cardiac tissue development and the progression of myocardial infarction this study investigated the expression levels of the MEIS1, HIRA, and Myocardin genes in premature CAD patients compared to healthy subjects and evaluated the relationships between these genes and possible inflammatory factors. METHODS AND RESULTS The study conducted a case-control design involving 35 CAD patients and 35 healthy individuals. Peripheral blood mononuclear cells (PBMCs) were collected, and gene expression analysis was performed using real-time PCR. Compared with control group, the number of PBMCs in the CAD group exhibited greater MEIS1 and HIRA gene expression, with fold changes of 2.45 and 3.6. The expression of MEIS1 exhibited a negative correlation with IL-10 (r= -0.312) expression and positive correlation with Interleukin (IL)-6 (r = 0.415) and tumor necrosis factor (TNF)-α (r = 0.534) gene expression. Moreover, there was an inverse correlation between the gene expression of HIRA and that of IL-10 (r= -0.326), and a positive correlation was revealed between the expression of this gene and that of the IL-6 (r = 0.453) and TNF-α (r = 0.572) genes. CONCLUSION This research demonstrated a disparity in expression levels of MEIS1, HIRA, and Myocardin, between CAD and healthy subjects. The results showed that, MEIS1 and HIRA play significant roles in regulating the synthesis of proinflammatory cytokines, namely, TNF-α and IL-6.
Collapse
Affiliation(s)
- Elnaz Javanshir
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | | | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Erfan Banisefid
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Neda Roshanravan
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
6
|
Expression characteristics and interaction networks of microRNAs in spleen tissues of grass carp (Ctenopharyngodon idella). PLoS One 2022; 17:e0266189. [PMID: 35344574 PMCID: PMC8959171 DOI: 10.1371/journal.pone.0266189] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
The spleen is an important immune organ in fish. MicroRNAs (miRNAs) have been shown to play an important role in the regulation of immune function. However, miRNA expression profiles and their interaction networks associated with the postnatal late development of spleen tissue are still poorly understood in fish. The grass carp (Ctenopharyngodon idella) is an important economic aquaculture species in China. Here, two small RNA libraries were constructed from the spleen tissue of healthy grass carp at one-year-old and three-year-old. A total of 324 known conserved miRNAs and 9 novel miRNAs were identified by using bioinformatic analysis. Family analysis showed that 23 families such as let-7, mir-1, mir-10, mir-124, mir-8, mir-7, mir-9, and mir-153 were highly conserved between vertebrates and invertebrates. In addition, 14 families such as mir-459, mir-430, mir-462, mir-7147, mir-2187, and mir-722 were present only in fish. Expression analysis showed that the expression patterns of miRNAs in the spleen of one-year-old and three-year-old grass carp were highly consistent, and the percentage of miRNAs with TPM > 100 was above 39%. Twenty significant differentially expressed (SDE) miRNAs were identified. Gene ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis showed that these SDE miRNAs were primarily involved in erythrocyte differentiation, lymphoid organ development, immune response, lipid metabolic process, the B cell receptor signaling pathway, the T cell receptor signaling pathway, and the PPAR signaling pathway. In addition, the following miRNA-mRNA interaction networks were constructed: immune and hematopoietic, cell proliferation and differentiation, and lipid metabolism. This study determined the miRNA transcriptome as well as miRNA-mRNA interaction networks in normal spleen tissue during the late development stages of grass carp. The results expand the number of known miRNAs in grass carp and are a valuable resource for better understanding the molecular biology of the spleen development in grass carp.
Collapse
|
7
|
Transcription factor Meis1 act as a new regulator of ischemic arrhythmias in mice. J Adv Res 2021; 39:275-289. [PMID: 35777912 PMCID: PMC9263651 DOI: 10.1016/j.jare.2021.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/25/2021] [Accepted: 11/10/2021] [Indexed: 11/20/2022] Open
Abstract
The reduction of Meis1 after MI leads to an increased susceptibility to arrhythmia. Meis1 deficiency is related to ubiquitination proteasome pathway mediated by CDC20. Meis1 acts as a new transcription activator for SCN5A in cardiomyocytes. After Meis1 recovery, the electrophysiological function in cardiomyocytes are improved. Meis1 is a new target for the treatment of arrhythmia after myocardial infarction.
Introduction Objectives Methods Results Conclusion
Collapse
|
8
|
Yao M, Gu Y, Yang Z, Zhong K, Chen Z. MEIS1 and its potential as a cancer therapeutic target (Review). Int J Mol Med 2021; 48:181. [PMID: 34318904 PMCID: PMC8354308 DOI: 10.3892/ijmm.2021.5014] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 07/08/2021] [Indexed: 01/26/2023] Open
Abstract
Meis homeobox 1 (Meis1) was initially discovered in 1995 as a factor involved in leukemia in an animal model. Subsequently, 2 years later, MEIS1, the human homolog, was cloned in the liver and cerebellum, and was found to be highly expressed in myeloid leukemia cells. The MEIS1 gene, located on chromosome 2p14, encodes a 390-amino acid protein with six domains. The expression of homeobox protein MEIS1 is affected by cell type, age and environmental conditions, as well as the pathological state. Certain types of modifications of MEIS1 and its protein interaction with homeobox or pre-B-cell leukemia homeobox proteins have been described. As a transcription factor, MEIS1 protein is involved in cell proliferation in leukemia and some solid tumors. The present review article discusses the molecular biology, modifications, protein-protein interactions, as well as the role of MEIS1 in cell proliferation of cancer cells and MEIS1 inhibitors. It is suggested by the available literature MEIS1 has potential to become a cancer therapeutic target.
Collapse
Affiliation(s)
- Maozhong Yao
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Yong Gu
- Clinical Research Center, Hainan Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Haikou, Hainan 570203, P.R. China
| | - Zhaoxin Yang
- Teaching Experimental Animal Center, Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Keyan Zhong
- Teaching Experimental Animal Center, Research Center for Drug Safety Evaluation of Hainan Province, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| | - Zhanjuan Chen
- Chemical Experiment Teaching Center, College of Pharmacy, Hainan Medical University, Haikou, Hainan 571199, P.R. China
| |
Collapse
|
9
|
Parial R, Li H, Li J, Archacki S, Yang Z, Wang IZ, Chen Q, Xu C, Wang QK. Role of epigenetic m 6 A RNA methylation in vascular development: mettl3 regulates vascular development through PHLPP2/mTOR-AKT signaling. FASEB J 2021; 35:e21465. [PMID: 33788967 DOI: 10.1096/fj.202000516rr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 11/11/2022]
Abstract
N6 -methyladenosine (m6A) methylation is the most prevalent RNA modification, and it emerges as an important regulatory mechanism of gene expression involved in many cellular and biological processes. However, the role of m6 A methylation in vascular development is not clear. The m6 A RNA methylation is regulated by dynamic interplay among methyltransferases, binding proteins, and demethylases. Mettl3 is a member of the mettl3-mettl14 methyltransferase complex, referred to as writers that catalyze m6A RNA methylation. Here, we used CRISPR-Cas9 genome editing to develop two lines of knockout (KO) zebrafish for mettl3. Heterozygous mettl3+/- KO embryos show defective vascular development, which is directly visible in fli-EGFP and flk-EGFP zebrafish. Alkaline phosphatase staining and whole mount in situ hybridization with cdh5, and flk markers demonstrated defective development of intersegmental vessels (ISVs), subintestinal vessels (SIVs), interconnecting vessels (ICVs) and dorsal longitudinal anastomotic vessels (DLAV) in both heterozygous mettl3+/- and homozygous mettl3-/- KO zebrafish embryos. Similar phenotypes were observed in zebrafish embryos with morpholino knockdown (KD) of mettl3; however, the vascular defects were rescued fully by overexpression of constitutively active AKT1. KD of METTL3 in human endothelial cells inhibited cell proliferation, migration, and capillary tube formation. Mechanistically, mettl3 KO and KD significantly reduced the levels of m6 A RNA methylation, and AKT phosphorylation (S473) by an increase in the expression of phosphatase enzyme PHLPP2 and reduction in the phosphorylation of mTOR (S2481), a member of the phosphatidylinositol 3-kinase-related kinase family of protein kinases. These data suggest that m6 A RNA methylation regulates vascular development via PHLPP2/mTOR-AKT signaling.
Collapse
Affiliation(s)
- Ramendu Parial
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Hui Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Jia Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Stephen Archacki
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Zhongcheng Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Isabel Z Wang
- Symbolic Systems Program, Stanford University, Stanford, CA, USA
| | - Qiuyun Chen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P.R. China
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology and Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, P.R. China.,Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.,Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| |
Collapse
|
10
|
Luo C, Tang B, Qin S, Yuan C, Du Y, Yang J. GATA2 regulates the CAD susceptibility gene ADTRP rs6903956 through preferential interaction with the G allele. Mol Genet Genomics 2021; 296:799-808. [PMID: 33856550 DOI: 10.1007/s00438-021-01782-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 03/29/2021] [Indexed: 02/06/2023]
Abstract
Myocardial infarction (MI) is a frequent outcome of coronary artery disease (CAD) and the key factor contributing to worldwide disability and death. Genetic factors contribute to the pathogenesis of CAD/MI, and SNP rs6903956 in the ADTRP gene was first found associated with CAD/MI in the Chinese Han population, which was successfully replicated in other cohorts. However, whether rs6903956 is a functional SNP and its risk mechanism to CAD/MI remains unknown. The ADTRP gene-encoded androgen-dependent TFPI regulating protein regulates vascular endothelial cell function, endothelial-monocyte adhesion, and thrombosis. The allele A of rs6903956, in particular, is associated with lower ADTRP mRNA levels in lymphocytes. In the current study, we found that SNP rs6903956 exhibits allelic differences in transcriptional activity by interacting with GATA2. Also, the A allele conferred a greater risk of CAD and MI, lowered transcriptional activity, and GATA2 binding ability as compared to the G allele. Our findings provide details on how rs6903956 regulates the expression of ADTRP and may provide novel insights into CAD pathology and susceptibility.
Collapse
Affiliation(s)
- Chunyan Luo
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, No.8, Da Xue Road, Yichang, 443002, Hubei Province, People's Republic of China. .,The Institute of Infection and Inflammation, China Three Gorges University, Yichang, 443002, Hubei, China.
| | - Bo Tang
- Department of Pharmacology, Institute of Material Medical, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Subo Qin
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Chengfu Yuan
- Department of Biochemistry, China Three Gorges University, Yichang, 443002, Hubei, China
| | - Youqin Du
- Department of Microbiology and Immunology, Medical College, China Three Gorges University, No.8, Da Xue Road, Yichang, 443002, Hubei Province, People's Republic of China
| | - Jian Yang
- Department of Cardiology, The People's Hospital of China Three Gorges University, Yichang, 443000, Hubei Province, China.
| |
Collapse
|
11
|
Guo Z, Mo Z. Regulation of endothelial cell differentiation in embryonic vascular development and its therapeutic potential in cardiovascular diseases. Life Sci 2021; 276:119406. [PMID: 33785330 DOI: 10.1016/j.lfs.2021.119406] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 03/05/2021] [Accepted: 03/14/2021] [Indexed: 12/17/2022]
Abstract
During vertebrate development, the cardiovascular system begins operating earlier than any other organ in the embryo. Endothelial cell (EC) forms the inner lining of blood vessels, and its extensive proliferation and migration are requisite for vasculogenesis and angiogenesis. Many aspects of cellular biology are involved in vasculogenesis and angiogenesis, including the tip versus stalk cell specification. Recently, epigenetics has attracted growing attention in regulating embryonic vascular development and controlling EC differentiation. Some proteins that regulate chromatin structure have been shown to be directly implicated in human cardiovascular diseases. Additionally, the roles of important EC signaling such as vascular endothelial growth factor and its receptors, angiopoietin-1 and tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-2, and transforming growth factor-β in EC differentiation during embryonic vasculature development are briefly discussed in this review. Recently, the transplantation of human induced pluripotent stem cell (iPSC)-ECs are promising approaches for the treatment of ischemic cardiovascular disease including myocardial infarction. Patient-specific iPSC-derived EC is a potential new target to study differences in gene expression or response to drugs. However, clinical application of the iPSC-ECs in regenerative medicine is often limited by the challenges of maintaining cell viability and function. Therefore, novel insights into the molecular mechanisms underlying EC differentiation might provide a better understanding of embryonic vascular development and bring out more effective EC-based therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Zi Guo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhaohui Mo
- Department of Endocrinology, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
12
|
Yuan F, Yun Y, Fan H, Li Y, Lu L, Liu J, Feng W, Chen SY. MicroRNA-135a Protects Against Ethanol-Induced Apoptosis in Neural Crest Cells and Craniofacial Defects in Zebrafish by Modulating the Siah1/p38/p53 Pathway. Front Cell Dev Biol 2020; 8:583959. [PMID: 33134300 PMCID: PMC7561719 DOI: 10.3389/fcell.2020.583959] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/14/2020] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) are small non-coding RNAs that are involved in various biological processes, including apoptosis, by regulating gene expression. This study was designed to test the hypothesis that ethanol-induced downregulation of miR-135a contributes to ethanol-induced apoptosis in neural crest cells (NCCs) by upregulating Siah1 and activating the p38 mitogen-activated protein kinase (MAPK)/p53 pathway. We found that treatment with ethanol resulted in a significant decrease in miR-135a expression in both NCCs and zebrafish embryos. Ethanol-induced downregulation of miR-135a resulted in the upregulation of Siah1 and the activation of the p38 MAPK/p53 pathway and increased apoptosis in NCCs and zebrafish embryos. Ethanol exposure also resulted in growth retardation and developmental defects that are characteristic of fetal alcohol spectrum disorders (FASD) in zebrafish. Overexpression of miRNA-135a significantly reduced ethanol-induced upregulation of Siah1 and the activation of the p38 MAPK/p53 pathway and decreased ethanol-induced apoptosis in NCCs and zebrafish embryos. In addition, ethanol-induced growth retardation and craniofacial defects in zebrafish larvae were dramatically diminished by the microinjection of miRNA-135a mimics. These results demonstrated that ethanol-induced downregulation of miR-135a contributes to ethanol-induced apoptosis in NCCs by upregulating Siah1 and activating the p38 MAPK/p53 pathway and that the overexpression of miRNA-135a can protect against ethanol-induced apoptosis in NCCs and craniofacial defects in a zebrafish model of FASD.
Collapse
Affiliation(s)
- Fuqiang Yuan
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Yang Yun
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States.,College of Environment and Resource, Research Center of Environment and Health, Shanxi University, Taiyuan, China
| | - Huadong Fan
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Yihong Li
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Lanhai Lu
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Jie Liu
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| | - Wenke Feng
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States.,Department of Medicine, University of Louisville, Louisville, KY, United States
| | - Shao-Yu Chen
- Department of Pharmacology and Toxicology, University of Louisville Health Sciences Center, Louisville, KY, United States.,University of Louisville Alcohol Research Center, Louisville, KY, United States
| |
Collapse
|
13
|
Johnson SC, Chakraborty S, Drosou A, Cunnea P, Tzovaras D, Nixon K, Zawieja DC, Muthuchamy M, Fotopoulou C, Moore JE. Inflammatory state of lymphatic vessels and miRNA profiles associated with relapse in ovarian cancer patients. PLoS One 2020; 15:e0230092. [PMID: 32716937 PMCID: PMC7384632 DOI: 10.1371/journal.pone.0230092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 07/05/2020] [Indexed: 01/20/2023] Open
Abstract
Lymphogenic spread is associated with poor prognosis in epithelial ovarian cancer (EOC), yet little is known regarding roles of non-peri-tumoural lymphatic vessels (LVs) outside the tumour microenvironment that may impact relapse. The aim of this feasibility study was to assess whether inflammatory status of the LVs and/or changes in the miRNA profile of the LVs have potential prognostic and predictive value for overall outcome and risk of relapse. Samples of macroscopically normal human lymph LVs (n = 10) were isolated from the external iliac vessels draining the pelvic region of patients undergoing debulking surgery. This was followed by quantification of the inflammatory state (low, medium and high) and presence of cancer-infiltration of each LV using immunohistochemistry. LV miRNA expression profiling was also performed, and analysed in the context of high versus low inflammation, and cancer-infiltrated versus non-cancer-infiltrated. Results were correlated with clinical outcome data including relapse with an average follow-up time of 13.3 months. The presence of a high degree of inflammation correlated significantly with patient relapse (p = 0.033). Cancer-infiltrated LVs showed a moderate but non-significant association with relapse (p = 0.07). Differential miRNA profiles were identified in cancer-infiltrated LVs and those with high versus low inflammation. In particular, several members of the let-7 family were consistently down-regulated in highly inflamed LVs (>1.8-fold, p<0.05) compared to the less inflamed ones. Down-regulation of the let-7 family appears to be associated with inflammation, but whether inflammation contributes to or is an effect of cancer-infiltration requires further investigation.
Collapse
Affiliation(s)
- Sarah C. Johnson
- Department of Bioengineering, Imperial College London, London, United Kingdom
| | | | - Anastasios Drosou
- Information Technologies Institute Centre for Research & Technology Hellas, Thessaloniki, Greece
| | - Paula Cunnea
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - Dimitrios Tzovaras
- Information Technologies Institute Centre for Research & Technology Hellas, Thessaloniki, Greece
| | - Katherine Nixon
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - David C. Zawieja
- College of Medicine, Texas A&M University, TX, United States of America
| | | | - Christina Fotopoulou
- Department of Surgery and Cancer, Imperial College London, London, United Kingdom
| | - James E. Moore
- Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
14
|
Balasubramanian S, Raghunath A, Perumal E. Role of epigenetics in zebrafish development. Gene 2019; 718:144049. [DOI: 10.1016/j.gene.2019.144049] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
|
15
|
Portrait of Tissue-Specific Coexpression Networks of Noncoding RNAs (miRNA and lncRNA) and mRNAs in Normal Tissues. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2019; 2019:9029351. [PMID: 31565069 PMCID: PMC6745163 DOI: 10.1155/2019/9029351] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 08/01/2019] [Accepted: 08/10/2019] [Indexed: 02/01/2023]
Abstract
Genes that encode proteins playing a role in more than one biological process are frequently dependent on their tissue context, and human diseases result from the altered interplay of tissue- and cell-specific processes. In this work, we performed a computational approach that identifies tissue-specific co-expression networks by integrating miRNAs, long-non-coding RNAs, and mRNAs in more than eight thousands of human samples from thirty normal tissue types. Our analysis (1) shows that long-non coding RNAs and miRNAs have a high specificity, (2) confirms several known tissue-specific RNAs, and (3) identifies new tissue-specific co-expressed RNAs that are currently still not described in the literature. Some of these RNAs interact with known tissue-specific RNAs or are crucial in key cancer functions, suggesting that they are implicated in tissue specification or cell differentiation.
Collapse
|
16
|
Schulte D, Geerts D. MEIS transcription factors in development and disease. Development 2019; 146:146/16/dev174706. [PMID: 31416930 DOI: 10.1242/dev.174706] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/28/2019] [Indexed: 12/12/2022]
Abstract
MEIS transcription factors are key regulators of embryonic development and cancer. Research on MEIS genes in the embryo and in stem cell systems has revealed novel and surprising mechanisms by which these proteins control gene expression. This Primer summarizes recent findings about MEIS protein activity and regulation in development, and discusses new insights into the role of MEIS genes in disease, focusing on the pathogenesis of solid cancers.
Collapse
Affiliation(s)
- Dorothea Schulte
- Institute of Neurology (Edinger Institute), University Hospital Frankfurt, Goethe University, 60528 Frankfurt, Germany
| | - Dirk Geerts
- Department of Medical Biology L2-109, Amsterdam University Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
17
|
Ji C, Guo X, Ren J, Zu Y, Li W, Zhang Q. Transcriptomic analysis of microRNAs-mRNAs regulating innate immune response of zebrafish larvae against Vibrio parahaemolyticus infection. FISH & SHELLFISH IMMUNOLOGY 2019; 91:333-342. [PMID: 31129189 DOI: 10.1016/j.fsi.2019.05.050] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 06/09/2023]
Abstract
In recent years, microRNAs (miRNAs) have been shown to play important roles in immunity. Analyses of the functions of miRNAs and their targets are useful in understanding the regulation of the immune response. To understand the relationships between miRNAs and their targets during infection, we used zebrafish as an infection model in which to characterize the miRNA and mRNA transcriptomes of zebrafish larvae infected with Vibrio parahaemolyticus. We identified the differentially expressed miRNAs and mRNAs. Overall, 37 known zebrafish miRNAs were differentially expressed in the infection group and 107 predicted target genes of 26 miRNAs were differentially expressed in the mRNA transcriptome. These targets with specific Gene Ontology (GO) terms, such as peripheral nervous system neuron axonogenesis, organophosphate metabolic process, heme binding, protein binding, tetrapyrrole binding, protein dimerization activity, and aromatase activity, which regulate nerve conduction, energy metabolism, hematopoiesis, and protein synthesis. They were also associated with Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways such as phototransduction, tryptophan metabolism, notch signaling, and purine metabolism. Our findings indicate that miRNAs regulate the innate immune response via complex networks, and zebrafish (Danio rerio, dre)-miR-205-3p, dre-miR-141-5p, dre-miR-200a-5p, dre-miR-92a-2-5p, dre-miR-192, and dre-miR-1788 may play important roles in the innate immune response by regulating target genes.
Collapse
Affiliation(s)
- Ce Ji
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Xinya Guo
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Jianfeng Ren
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Yao Zu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China
| | - Weiming Li
- Department of Fisheries and Wildlife, Michigan State University, East Lansing, MI, 48824, USA
| | - Qinghua Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai, 201306, China; International Research Center for Marine Biosciences, Ministry of Science and Technology, Shanghai Ocean University, Shanghai, 201306, China; Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, 201306, China.
| |
Collapse
|
18
|
Lareyre F, Clément M, Moratal C, Loyer X, Jean-Baptiste E, Hassen-Khodja R, Chinetti G, Mallat Z, Raffort J. Differential micro-RNA expression in diabetic patients with abdominal aortic aneurysm. Biochimie 2019; 162:1-7. [DOI: 10.1016/j.biochi.2019.03.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 03/18/2019] [Indexed: 12/29/2022]
|
19
|
Jiang L, Meng W, Yu G, Yin C, Wang Z, Liao L, Meng F. MicroRNA-144 targets APP to regulate AML1/ETO + leukemia cell migration via the p-ERK/c-Myc/MMP-2 pathway. Oncol Lett 2019; 18:2034-2042. [PMID: 31423275 DOI: 10.3892/ol.2019.10477] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 12/04/2018] [Indexed: 12/30/2022] Open
Abstract
Extramedullary infiltration (EMI) is common in patients with acute myeloid leukemia (AML) and is closely associated with the prognosis of disease. We previously reported that patients carrying the AML1/ETO (A/E) fusion gene and expressing the amyloid precursor protein (APP) tended to develop EMI, and had a poor prognosis. In the present study, the relapse-free survival (RFS) time and overall survival (OS) time were significantly lower in patients with EMI. The results demonstrated that the EMI incidence was significantly higher (P<0.05), while the RFS and OS rates were significantly lower (P<0.05), in patients with high APP expression. Kasumi-1 cells, which are A/E+, and the APP gene were used as the in vitro cell model to detect the mechanism of action in detail. Following the knockdown of APP expression, cell migration was significantly reduced (P<0.05). Furthermore, western blotting demonstrated that the protein expression of phosphorylated extracellular-signal-regulated kinase (p-ERK), matrix metalloproteinase-2 (MMP-2) and c-Myc was markedly reduced following interference of APP, while the expression of CXCR4 and MMP-9 was not altered. Kasumi-1 cells were co-cultured with p-ERK or c-Myc inhibitors and demonstrated that the APP/p-ERK/c-Myc/MMP-2 pathway was involved in signal transduction and regulation of cell migration. MicroRNA-144 (miR-144) mimics and transfected Kasumi-1 cells were generated. Reverse transcription-quantitative polymerase chain reaction and western blotting demonstrated that miR-144 was a negative regulator of APP. Taken together, the findings of the present study suggest that miR-144 negatively targets the APP gene and regulates cell migration via the APP/p-ERK/c-Myc/MMP-2 pathway.
Collapse
Affiliation(s)
- Ling Jiang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Guopan Yu
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Changxin Yin
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Zhixiang Wang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Libin Liao
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510500, P.R. China
| | - Fanyi Meng
- Department of Hematology, Kang Hua Hospital, Dongguan, Guangdong 523080, P.R. China
| |
Collapse
|
20
|
Delineating the Dynamic Transcriptome Response of mRNA and microRNA during Zebrafish Heart Regeneration. Biomolecules 2018; 9:biom9010011. [PMID: 30597924 PMCID: PMC6359357 DOI: 10.3390/biom9010011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/19/2018] [Accepted: 12/21/2018] [Indexed: 12/12/2022] Open
Abstract
Heart diseases are the leading cause of death for the vast majority of people around the world, which is often due to the limited capability of human cardiac regeneration. In contrast, zebrafish have the capacity to fully regenerate their hearts after cardiac injury. Understanding and activating these mechanisms would improve health in patients suffering from long-term consequences of ischemia. Therefore, we monitored the dynamic transcriptome response of both mRNA and microRNA in zebrafish at 1–160 days post cryoinjury (dpi). Using a control model of sham-operated and healthy fish, we extracted the regeneration specific response and further delineated the spatio-temporal organization of regeneration processes such as cell cycle and heart function. In addition, we identified novel (miR-148/152, miR-218b and miR-19) and previously known microRNAs among the top regulators of heart regeneration by using theoretically predicted target sites and correlation of expression profiles from both mRNA and microRNA. In a cross-species effort, we validated our findings in the dynamic process of rat myoblasts differentiating into cardiomyocytes-like cells (H9c2 cell line). Concluding, we elucidated different phases of transcriptomic responses during zebrafish heart regeneration. Furthermore, microRNAs showed to be important regulators in cardiomyocyte proliferation over time.
Collapse
|
21
|
Wang X, Li J, Yang Z, Wang L, Li L, Deng W, Zhou J, Wang L, Xu C, Chen Q, Wang QK. phlda3 overexpression impairs specification of hemangioblasts and vascular development. FEBS J 2018; 285:4071-4081. [PMID: 30188605 PMCID: PMC6218282 DOI: 10.1111/febs.14653] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 08/20/2018] [Accepted: 09/04/2018] [Indexed: 01/25/2023]
Abstract
The phlda3 gene encodes a small, 127-amino acid protein with only a PH domain, and is involved in tumor suppression, proliferation of islet β-cells, insulin secretion, glucose tolerance, and liver injury. However, the role of phlda3 in vascular development is unknown. Here, we show that phlda3 overexpression decreases the expression levels of hemangioblast markers scl, fli1, and etsrp and intersegmental vessel (ISV) markers flk1 and cdh5, and disrupts ISV development in tg(flk1:GFP) and tg(fli1:GFP) zebrafish. Moreover, phlda3 overexpression inhibits the activation of protein kinase B (AKT) in zebrafish embryos, and the developmental defects of ISVs by phlda3 overexpression were reversed by the expression of a constitutively active form of AKT. These data suggest that phlda3 is a negative regulator of hemangioblast specification and ISV development via AKT signaling.
Collapse
Affiliation(s)
- Xiaojing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Jia Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Zhongcheng Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Li Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Lei Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Wenqing Deng
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Juan Zhou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Longfei Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic; Department of Molecular Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Qing K Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-X Institute, Huazhong University of Science and Technology, Wuhan, P. R. China
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic; Department of Molecular Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
22
|
Chan J, Hu X, Wang C, Xu Q. miRNA-152 targets GATA1 to regulate erythropoiesis in Chionodraco hamatus. Biochem Biophys Res Commun 2018; 501:711-717. [DOI: 10.1016/j.bbrc.2018.05.053] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Accepted: 05/09/2018] [Indexed: 12/12/2022]
|
23
|
Pinhal D, Bovolenta LA, Moxon S, Oliveira AC, Nachtigall PG, Acencio ML, Patton JG, Hilsdorf AWS, Lemke N, Martins C. Genome-wide microRNA screening in Nile tilapia reveals pervasive isomiRs' transcription, sex-biased arm switching and increasing complexity of expression throughout development. Sci Rep 2018; 8:8248. [PMID: 29844338 PMCID: PMC5974277 DOI: 10.1038/s41598-018-26607-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
MicroRNAs (miRNAs) are key regulators of gene expression in multicellular organisms. The elucidation of miRNA function and evolution depends on the identification and characterization of miRNA repertoire of strategic organisms, as the fast-evolving cichlid fishes. Using RNA-seq and comparative genomics we carried out an in-depth report of miRNAs in Nile tilapia (Oreochromis niloticus), an emergent model organism to investigate evo-devo mechanisms. Five hundred known miRNAs and almost one hundred putative novel vertebrate miRNAs have been identified, many of which seem to be teleost-specific, cichlid-specific or tilapia-specific. Abundant miRNA isoforms (isomiRs) were identified with modifications in both 5p and 3p miRNA transcripts. Changes in arm usage (arm switching) of nine miRNAs were detected in early development, adult stage and even between male and female samples. We found an increasing complexity of miRNA expression during ontogenetic development, revealing a remarkable synchronism between the rate of new miRNAs recruitment and morphological changes. Overall, our results enlarge vertebrate miRNA collection and reveal a notable differential ratio of miRNA arms and isoforms influenced by sex and developmental life stage, providing a better picture of the evolutionary and spatiotemporal dynamics of miRNAs.
Collapse
Affiliation(s)
- Danillo Pinhal
- Department of Genetics, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil.
| | - Luiz A Bovolenta
- Department of Physics and Biophysics, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Simon Moxon
- School of Biological Sciences, University of East Anglia (UEA), Norwich Research Park, Norwich, United Kingdom
| | - Arthur C Oliveira
- Department of Genetics, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Pedro G Nachtigall
- Department of Genetics, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Marcio L Acencio
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - James G Patton
- Stevenson Center, Vanderbilt University, Nashville, TN, USA
| | | | - Ney Lemke
- Department of Physics and Biophysics, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Cesar Martins
- Department of Morphology, Institute of Biosciences of Botucatu, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| |
Collapse
|
24
|
MEIS-1 level in unresectable hepatocellular carcinoma can predict the post-treatment outcomes of radiofrequency ablation. Oncotarget 2018; 9:15252-15265. [PMID: 29632641 PMCID: PMC5880601 DOI: 10.18632/oncotarget.24165] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 12/01/2017] [Indexed: 02/06/2023] Open
Abstract
Radiofrequency ablation (RFA) is a local-ablative therapy for unresectable hepatocellular carcinoma (HCC). At present, there is no predictive marker for RFA treatment outcomes. This work aimed to valuate myeloid ecotropic viral integration site 1 (MEIS-1) in predicting post-RFA treatment outcomes of unresectable HCC patients. The time to progression (TTP) and overall survival (OS) of 81 HCC patients who received RFA treatment were measured. The protein level of MEIS-1 in tumor specimens was measured by western blot. The role of MEIS-1 in RFA-treating HCC in vivo growth nude mouse model was examined via PET/CT imaging. Higher level of MEIS-1 in tumor tissue is associated with better RFA treatment outcomes. The median TTP was 9.0 (95% confidence interval [CI]: 6.8–11.3) months in patients with high MEIS-1 expression (n = 43) versus 6.0 (95% CI: 4.6–7.4) months in patients with low MEIS-1 expression (n = 38). Moreover, in rodent HCC model we found overexpression of MEIS-1 enhanced the anti-tumor effect of RFA treatment. We conclude that high level of MEIS-1 expression predicts better RFA treatment outcome in HCC.
Collapse
|
25
|
Wang L, Wang X, Wang L, Yousaf M, Li J, Zuo M, Yang Z, Gou D, Bao B, Li L, Xiang N, Jia H, Xu C, Chen Q, Wang QK. Identification of a new adtrp1-tfpi regulatory axis for the specification of primitive myelopoiesis and definitive hematopoiesis. FASEB J 2017; 32:183-194. [PMID: 28877957 DOI: 10.1096/fj.201700166rr] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 08/21/2017] [Indexed: 12/13/2022]
Abstract
A genomic variant in the human ADTRP [androgen-dependent tissue factor (TF) pathway inhibitor (TFPI) regulating protein] gene increases the risk of coronary artery disease, the leading cause of death worldwide. TFPI is the TF pathway inhibitor that is involved in coagulation. Here, we report that adtrp and tfpi form a regulatory axis that specifies primitive myelopoiesis and definitive hematopoiesis, but not primitive erythropoiesis or vasculogenesis. In zebrafish, there are 2 paralogues for adtrp (i.e., adtrp1 and adtrp2). Knockdown of adtrp1 expression inhibits the specification of hemangioblasts, as shown by decreased expression of the hemangioblast markers, etsrp, fli1a, and scl; blocks primitive hematopoiesis, as shown by decreased expression of pu.1, mpo, and l-plastin; and disrupts the specification of hematopoietic stem cells (definitive hematopoiesis), as shown by decreased expression of runx1 and c-myb However, adtrp1 knockdown does not affect erythropoiesis during primitive hematopoiesis (no effect on gata1 or h-bae1) or vasculogenesis (no effect on kdrl, ephb2a, notch3, dab2, or flt4). Knockdown of adtrp2 expression does not have apparent effects on all markers tested. Knockdown of adtrp1 reduced the expression of tfpi, and hematopoietic defects in adtrp1 morphants were rescued by tfpi overexpression. These data suggest that the regulation of tfpi expression is one potential mechanism by which adtrp1 regulates primitive myelopoiesis and definitive hematopoiesis.-Wang, L., Wang, X., Wang, L., Yousaf, M., Li, J., Zuo, M., Yang, Z., Gou, D., Bao, B., Li, L., Xiang, N., Jia, H., Xu, C., Chen, Q., Wang, Q. K. Identification of a new adtrp1-tfpi regulatory axis for the specification of primitive myelopoiesis and definitive hematopoiesis.
Collapse
Affiliation(s)
- Li Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojing Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Longfei Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Muhammad Yousaf
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Mengxia Zuo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Zhongcheng Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Dongzhi Gou
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Binghao Bao
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Xiang
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haibo Jia
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Chengqi Xu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; .,Department of Molecular Medicine, Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Qing Kenneth Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Cardio-X Center, College of Life Science and Technology, Center for Human Genome Research, Huazhong University of Science and Technology, Wuhan, China; .,Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Molecular Medicine, Lerner College of Medicine, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
26
|
Luo C, Pook E, Tang B, Zhang W, Li S, Leineweber K, Cheung SH, Chen Q, Bechem M, Hu JS, Laux V, Wang QK. Androgen inhibits key atherosclerotic processes by directly activating ADTRP transcription. Biochim Biophys Acta Mol Basis Dis 2017. [PMID: 28645652 DOI: 10.1016/j.bbadis.2017.06.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Low androgen levels are associated with an increased risk of coronary artery disease (CAD), thrombosis and myocardial infarction (MI), suggesting that androgen has a protective role. However, little is known about the underlying molecular mechanism. Our genome-wide association study identified the ADTRP gene encoding the androgen-dependent TFPI regulating protein as a susceptibility gene for CAD and MI. The expression level of ADTRP was regulated by androgen, but the molecular mechanism is unknown. In this study, we identified the molecular mechanism by which androgen regulates ADTRP expression and tested the hypothesis that androgen plays a protective role in cardiovascular disease by activating ADTRP expression. Luciferase assays with an ADTRP promoter luciferase reporter revealed that androgen regulated ADTRP transcription in a dose- and time-dependent manner, and the effect was abolished by three different androgen inhibitors, including pyrvinium pamoate, bicalutamide, and cyproterone acetate. Chromatin-immunoprecipitation showed that the androgen receptor bound to a half androgen response element (ARE, TGTTCT) located at +324bp from the ADTRP transcription start site. The ARE is required for concentration-dependent transcriptional activation of ADTRP. HL-60 monocyte adhesion to EAhy926 endothelial cells (ECs) and transmigration across the EC layer, the two processes critical to development of CAD and MI, were inhibited by androgen, but the effect was rescued by ADTRP siRNA and exacerbated by overexpression of ADTRP and its downstream genes PIK3R3 and MIA3. These data suggest that one molecular mechanism by which androgen confers protection against CAD is stimulation of ADTRP expression.
Collapse
Affiliation(s)
- Chunyan Luo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | | | - Bo Tang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | - Weiyi Zhang
- Bayer Healthcare Co Ltd, Innovation Center China, Beijing, PR China
| | - Sisi Li
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China
| | | | - Shing-Hu Cheung
- Bayer Healthcare Co Ltd, Innovation Center China, Beijing, PR China
| | - Qiuyun Chen
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44195, USA
| | | | - Jing-Shan Hu
- Bayer Healthcare Co Ltd, Innovation Center China, Beijing, PR China
| | - Volker Laux
- Bayer AG, Drug Discovery, 42096 Wuppertal, Germany.
| | - Qing Kenneth Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Center for Human Genome Research, Cardio-Institute, Huazhong University of Science and Technology, Wuhan 430074, PR China; Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44195, USA.
| |
Collapse
|
27
|
He J, Tian Y, Zhao Y, Liu Y, Tao Z, Li G, Niu D, Lu L, Lu Y. MiR-144 affects fatty acid composition by regulating ELOVL6 expression in duck hepatocytes. Cell Biol Int 2017; 41:691-696. [PMID: 28225172 DOI: 10.1002/cbin.10753] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Accepted: 02/19/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Jun He
- Institute of Animal Husbandry and Veterinary Science; Zhejiang Academy Agricultural Sciences; Hangzhou 310021 China
- Department of Animal Science; Wenzhou Vocational College of Science & Technology; Wenzhou 325006 China
| | - Yong Tian
- Institute of Animal Husbandry and Veterinary Science; Zhejiang Academy Agricultural Sciences; Hangzhou 310021 China
| | - Yan Zhao
- Department of Animal Science; Wenzhou Vocational College of Science & Technology; Wenzhou 325006 China
| | - Yali Liu
- Institute of Animal Husbandry and Veterinary Science; Zhejiang Academy Agricultural Sciences; Hangzhou 310021 China
| | - Zhengrong Tao
- Institute of Animal Husbandry and Veterinary Science; Zhejiang Academy Agricultural Sciences; Hangzhou 310021 China
| | - Guoqin Li
- Institute of Animal Husbandry and Veterinary Science; Zhejiang Academy Agricultural Sciences; Hangzhou 310021 China
| | - Dong Niu
- College of Animal Science; Zhejiang University; Hangzhou 310021 China
| | - Lizhi Lu
- Institute of Animal Husbandry and Veterinary Science; Zhejiang Academy Agricultural Sciences; Hangzhou 310021 China
| | - Yingru Lu
- Department of Emergency Medicine; First Affiliated Hospital of Wenzhou Medical University; Wenzhou 325000 China
| |
Collapse
|
28
|
Gao F, Liu P, Narayanan J, Yang M, Fish BL, Liu Y, Liang M, Jacobs ER, Medhora M. Changes in miRNA in the lung and whole blood after whole thorax irradiation in rats. Sci Rep 2017; 7:44132. [PMID: 28303893 PMCID: PMC5355888 DOI: 10.1038/srep44132] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 02/03/2017] [Indexed: 01/10/2023] Open
Abstract
We used a rat model of whole thorax x-ray irradiation to profile the microRNA (miRNA) in lung and blood up to 4 weeks after radiation. MiRNA from normal and irradiated Wistar rat lungs and whole blood were analyzed by next-generation sequencing and the changes by radiation were identified by differential deRNA-seq 1, 2, 3 and 4 weeks after irradiation. The average total reads/library was 2,703,137 with a mean of 88% mapping to the rat genome. Detailed profiles of 100 of the most abundant miRNA in rat blood and lung are described. We identified upregulation of 4 miRNA, miR-144-5p, miR-144-3p, miR-142-5p and miR-19a-3p in rat blood 2 weeks after radiation that have not previously been shown to be altered after radiation to the lung. Ingenuity Pathway Analysis identified signaling of inflammatory response pathways. These findings will support development of early detection methods, as well as mechanism(s) of injury and mitigation in patients after radiotherapy or radiological accidents.
Collapse
Affiliation(s)
- Feng Gao
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Pengyuan Liu
- Department of Physiology, Medical College of Wisconsin,Milwaukee, WI, USA
| | - Jayashree Narayanan
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Meiying Yang
- Department of Anesthesiology, Medical College of Wisconsin,Milwaukee, WI, USA
| | - Brian L Fish
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Yong Liu
- Department of Physiology, Medical College of Wisconsin,Milwaukee, WI, USA
| | - Mingyu Liang
- Department of Physiology, Medical College of Wisconsin,Milwaukee, WI, USA
| | - Elizabeth R Jacobs
- Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Research Service, Department of Veterans Affairs, Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| | - Meetha Medhora
- Department of Radiation Oncology, Medical College of Wisconsin, Milwaukee, WI, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, WI, USA.,Research Service, Department of Veterans Affairs, Zablocki Veterans Affairs Medical Center, Milwaukee, WI, USA
| |
Collapse
|
29
|
Zhu J, Cui L, Xu A, Yin X, Li F, Gao J. MEIS1 inhibits clear cell renal cell carcinoma cells proliferation and in vitro invasion or migration. BMC Cancer 2017; 17:176. [PMID: 28270206 PMCID: PMC5341457 DOI: 10.1186/s12885-017-3155-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Accepted: 02/23/2017] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Myeloid ecotropic viral integration site 1 (MEIS1) protein plays a synergistic causative role in acute myeloid leukemia (AML). However, MEIS1 has also shown to be a potential tumor suppressor in some other cancers, such as non-small-cell lung cancer (NSCLC) and prostate cancer. Although multiple roles of MEIS1 in cancer development and progression have been identified, there is an urgent demand to discover more functions of this molecule for further therapeutic design. METHODS MEIS1 was overexpressed via adenovirus vector in clear cell renal cell carcinoma (ccRCC) cells. Western blot and real-time qPCR (quantitative Polymerase Chain Reaction) was performed to examine the protein and mRNA levels of MEIS1. Cell proliferation, survival, in vitro migration and invasion were tested by MTT, colony formation, soft-agar, transwell (in vitro invasion/migration) assays, and tumor in vivo growthwas measured on nude mice model. In addition, flow-cytometry analysis was used to detect cell cycle arrest or non-apoptotic cell death of ccRCC cells induced by MEIS1. RESULTS MEIS1 exhibits a decreased expression in ccRCC cell lines than that in non-tumor cell lines. MEIS1 overexpression inhibits ccRCC cells proliferation and induces G1/S arrest concomitant with marked reduction of G1/S transition regulators, Cyclin D1 and Cyclin A. Moreover, MEIS1-1 overexpression also induces non-apoptotic cell death of ccRCC cells via decreasing the levels of pro-survival regulators Survivin and BCL-2. Transwell migration assay (TMA) shows that MEIS1 attenuates in vitro invasion and migration of ccRCC cells with down-regulated epithelial-mesenchymal transition (EMT) process. Further, in nude mice model, MEIS1 inhibits the in vivo growth of Caki-1 cells. CONCLUSIONS By investigating the role of MEIS1 in ccRCC cells' survival, proliferation, anchorage-independent growth, cell cycle progress, apoptosis and metastasis, in the present work, we propose that MEIS1 may play an important role in clear cell renal cell carcinoma (ccRCC) development.
Collapse
Affiliation(s)
- Jie Zhu
- Department of Urology, Chinese PLA Medical School/Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
| | - Liang Cui
- Department of Urology, Chinese PLA Medical School/Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
- Department of Urology, Civil Aviation General Hospital/Civil Aviation Medical College of Peking University, Beijing, 100123 People’s Republic of China
| | - Axiang Xu
- Department of Urology, Chinese PLA Medical School/Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
| | - Xiaotao Yin
- Department of Urology, Chinese PLA Medical School/Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
| | - Fanglong Li
- Department of Urology, Chinese PLA Medical School/Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
| | - Jiangping Gao
- Department of Urology, Chinese PLA Medical School/Chinese PLA General Hospital, Beijing, 100853 People’s Republic of China
| |
Collapse
|
30
|
Mesenchymal Stem Cell-Derived Microvesicles Support Ex Vivo Expansion of Cord Blood-Derived CD34(+) Cells. Stem Cells Int 2016; 2016:6493241. [PMID: 27042183 PMCID: PMC4799819 DOI: 10.1155/2016/6493241] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 01/17/2016] [Indexed: 12/29/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are known to support the characteristic properties of hematopoietic stem and progenitor cells (HSPCs) in the bone marrow hematopoietic microenvironment. MSCs are used in coculture systems as a feeder layer for the ex vivo expansion of umbilical cord blood (CB) to increase the relatively low number of HSPCs in CB. Findings increasingly suggest that MSC-derived microvesicles (MSC-MVs) play an important role in the biological functions of their parent cells. We speculate that MSC-MVs may recapitulate the hematopoiesis-supporting effects of their parent cells. In the current study, we found MSC-MVs containing microRNAs that are involved in the regulation of hematopoiesis. We also demonstrated that MSC-MVs could improve the expansion of CB-derived mononuclear cells and CD34+ cells and generate a greater number of primitive progenitor cells in vitro. Additionally, when MSC-MVs were added to the CB-MSC coculture system, they could improve the hematopoiesis-supporting effects of MSCs. These findings highlight the role of MSC-MVs in the ex vivo expansion of CB, which may offer a promising therapeutic approach in CB transplantation.
Collapse
|
31
|
Cardiac sodium channel regulator MOG1 regulates cardiac morphogenesis and rhythm. Sci Rep 2016; 6:21538. [PMID: 26903377 PMCID: PMC4763225 DOI: 10.1038/srep21538] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Accepted: 01/26/2016] [Indexed: 01/09/2023] Open
Abstract
MOG1 was initially identified as a protein that interacts with the small GTPase Ran involved in transport of macromolecules into and out of the nucleus. In addition, we have established that MOG1 interacts with the cardiac sodium channel Nav1.5 and regulates cell surface trafficking of Nav1.5. Here we used zebrafish as a model system to study the in vivo physiological role of MOG1. Knockdown of mog1 expression in zebrafish embryos significantly decreased the heart rate (HR). Consistently, the HR increases in embryos with over-expression of human MOG1. Compared with wild type MOG1 or control EGFP, mutant MOG1 with mutation E83D associated with Brugada syndrome significantly decreases the HR. Interestingly, knockdown of mog1 resulted in abnormal cardiac looping during embryogenesis. Mechanistically, knockdown of mog1 decreases expression of hcn4 involved in the regulation of the HR, and reduces expression of nkx2.5, gata4 and hand2 involved in cardiac morphogenesis. These data for the first time revealed a novel role that MOG1, a nucleocytoplasmic transport protein, plays in cardiac physiology and development.
Collapse
|
32
|
Chandra V, Kim JJ, Mittal B, Rai R. MicroRNA aberrations: An emerging field for gallbladder cancer management. World J Gastroenterol 2016; 22:1787-1799. [PMID: 26855538 PMCID: PMC4724610 DOI: 10.3748/wjg.v22.i5.1787] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Revised: 11/12/2015] [Accepted: 12/21/2015] [Indexed: 02/06/2023] Open
Abstract
Gallbladder cancer (GBC) is infrequent but most lethal biliary tract malignancy characterized by an advanced stage diagnosis and poor survival rates attributed to absence of specific symptoms and effective treatment options. These necessitate development of early prognostic/predictive markers and novel therapeutic interventions. MicroRNAs (miRNAs) are small, non-coding RNA molecules that play a key role in tumor biology by functioning like tumor suppressor- or onco- genes and their aberrant expression are associated with the pathogenesis of several neoplasms with overwhelming clinical implications. Since miRNA signature is tissue specific, here, we focused on current data concerning the miRNAs abberations in GBC pathogenesis. In GBC, miRNAs with tumor suppressor activity (miR-135-5p, miR-335, miR-34a, miR-26a, miR-146b-5p, Mir-218-5p, miR-1, miR-145, mir-130a) were found downregulated, while those with oncogenic property (miR-20a, miR-182, mir-155) were upregulated. The expression profile of miRNAs was significantly associated with GBC prognosis and prediction, and forced over-expression/ inhibition of these miRNAs was shown to affect tumor growth and development. Further, differential expression of miRNAs in the blood samples of GBC patients suggest miRNAs as promising noninvasive biomarker. Thus, miRNAs represent potential candidate for GBC management, though many hurdles need to be overcome before miRNAs therapy can be clinically applied to GBC prevention and treatment.
Collapse
|
33
|
Cai H, Xue Y, Wang P, Wang Z, Li Z, Hu Y, Li Z, Shang X, Liu Y. The long noncoding RNA TUG1 regulates blood-tumor barrier permeability by targeting miR-144. Oncotarget 2015; 6:19759-79. [PMID: 26078353 PMCID: PMC4637319 DOI: 10.18632/oncotarget.4331] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/25/2015] [Indexed: 01/16/2023] Open
Abstract
Blood-tumor barrier (BTB) limits the delivery of chemotherapeutic agent to brain tumor tissues. Long non-coding RNAs (lncRNAs) have been shown to play critical regulatory roles in various biologic processes of tumors. However, the role of lncRNAs in BTB permeability is unclear. LncRNA TUG1 (taurine upregulated gene 1) was highly expressed in glioma vascular endothelial cells from glioma tissues. It also upregulated in glioma co-cultured endothelial cells (GEC) from BTB model in vitro. Knockdown of TUG1 increased BTB permeability, and meanwhile down-regulated the expression of the tight junction proteins ZO-1, occludin, and claudin-5. Both bioinformatics and luciferase reporter assays demonstrated that TUG1 influenced BTB permeability via binding to miR-144. Furthermore, Knockdown of TUG1 also down-regulated Heat shock transcription factor 2 (HSF2), a transcription factor of the heat shock transcription factor family, which was defined as a direct and functional downstream target of miR-144. HSF2 up-regulated the promoter activities and interacted with the promoters of ZO-1, occludin, and claudin-5 in GECs. In conclusion, our results indicate that knockdown of TUG1 increased BTB permeability via binding to miR-144 and then reducing EC tight junction protein expression by targeting HSF2. Thus, TUG1 may represent a useful future therapeutic target for enhancing BTB permeability.
Collapse
Affiliation(s)
- Heng Cai
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yixue Xue
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, People's Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, People's Republic of China
| | - Ping Wang
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, People's Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, People's Republic of China
| | - Zhenhua Wang
- Department of Physiology, College of Basic Medicine, China Medical University, Shenyang, People's Republic of China
| | - Zhen Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yi Hu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Zhiqing Li
- Department of Neurobiology, College of Basic Medicine, China Medical University, Shenyang, People's Republic of China
- Institute of Pathology and Pathophysiology, China Medical University, Shenyang, People's Republic of China
| | - Xiuli Shang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, People's Republic of China
| | - Yunhui Liu
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| |
Collapse
|
34
|
Huang C, Chen N, Wu X, Huang C, He Y, Tang R, Wang W, Wang H. The zebrafish miR‐462/miR‐731 cluster is induced under hypoxic stress
via
hypoxia‐inducible factor 1α and functions in cellular adaptations. FASEB J 2015; 29:4901-13. [DOI: 10.1096/fj.14-267104] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 08/03/2015] [Indexed: 12/13/2022]
Affiliation(s)
- Chun‐Xiao Huang
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
| | - Nan Chen
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
| | - Xin‐Jie Wu
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
| | - Cui‐Hong Huang
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
| | - Yan He
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
| | - Rong Tang
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
- Freshwater Aquaculture Collaborative Innovation Center of Hubei ProvinceWuhanHubeiChina
| | - Wei‐Min Wang
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
- Freshwater Aquaculture Collaborative Innovation Center of Hubei ProvinceWuhanHubeiChina
| | - Huan‐Ling Wang
- Key Laboratory of Freshwater Animal Breeding and Key Laboratory of Agricultural Animal GeneticsBreeding and Reproduction, Ministry of Education, College of FisheryHuazhong Agricultural UniversityWuhanHubeiChina
- Freshwater Aquaculture Collaborative Innovation Center of Hubei ProvinceWuhanHubeiChina
| |
Collapse
|
35
|
Yan L, Lee S, Lazzaro DR, Aranda J, Grant MB, Chaqour B. Single and Compound Knock-outs of MicroRNA (miRNA)-155 and Its Angiogenic Gene Target CCN1 in Mice Alter Vascular and Neovascular Growth in the Retina via Resident Microglia. J Biol Chem 2015; 290:23264-81. [PMID: 26242736 DOI: 10.1074/jbc.m115.646950] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Indexed: 01/09/2023] Open
Abstract
The response of the retina to ischemic insult typically leads to aberrant retinal neovascularization, a major cause of blindness. The epigenetic regulation of angiogenic gene expression by miRNAs provides new prospects for their therapeutic utility in retinal neovascularization. Here, we focus on miR-155, a microRNA functionally important in inflammation, which is of paramount importance in the pathogenesis of retinal neovascularization. Whereas constitutive miR-155-deficiency in mice results in mild vascular defects, forced expression of miR-155 causes endothelial hyperplasia and increases microglia count and activation. The mouse model of oxygen-induced retinopathy, which recapitulates ischemia-induced aberrant neovessel growth, is characterized by increased expression of miR-155 and localized areas of microglia activation. Interestingly, miR-155 deficiency in mice reduces microglial activation, curtails abnormal vessel growth, and allows for rapid normalization of the retinal vasculature following ischemic insult. miR-155 binds to the 3'-UTR and represses the expression of the CCN1 gene, which encodes an extracellular matrix-associated integrin-binding protein that both promotes physiological angiogenesis and harnesses growth factor-induced abnormal angiogenic responses. Single CCN1 deficiency or double CCN1 and miR-155 knock-out in mice causes retinal vascular malformations typical of faulty maturation, mimicking the vascular alterations of miR-155 gain of function. During development, the miR-155/CCN1 regulatory axis balances the proangiogenic and proinflammatory activities of microglia to allow for their function as guideposts for sprout fusion and anastomosis. Under ischemic conditions, dysregulated miR-155 and CCN1 expression increases the inflammatory load and microglial activation, prompting aberrant angiogenic responses. Thus, miR-155 functions in tandem with CCN1 to modulate inflammation-induced vascular homeostasis and repair.
Collapse
Affiliation(s)
- Lulu Yan
- From the Departments of Cell Biology
| | | | | | | | - Maria B Grant
- the Departments of Ophthalmology and Cellular and Integrative Physiology, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | - Brahim Chaqour
- From the Departments of Cell Biology, Ophthalmology, and the SUNY Eye Institute, SUNY Downstate Medical Center, Brooklyn, New York 11203 and
| |
Collapse
|
36
|
Zhu H, Leung SW. Identification of microRNA biomarkers in type 2 diabetes: a meta-analysis of controlled profiling studies. Diabetologia 2015; 58:900-11. [PMID: 25677225 DOI: 10.1007/s00125-015-3510-2] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Accepted: 01/12/2015] [Indexed: 02/08/2023]
Abstract
AIMS/HYPOTHESIS The aim was to identify potential microRNA (miRNA) biomarkers of type 2 diabetes. METHODS Controlled studies were retrieved from PubMed to compare miRNA expression profiles of type 2 diabetes and nondiabetic control samples. Meta-analysis under a random effects model was conducted. Subgroup analyses examined tissue specificity and species specificity. Sensitivity analyses were also performed to explain the heterogeneity among studies. Results were represented as log odds ratios (logOR), 95% confidence intervals (CI) and p values after Bonferroni correction. RESULTS Among 343 differentially expressed miRNAs in 38 miRNA expression profiling studies published between 1993 and March 2014, only 151 miRNAs were tested by multiple studies, out of which 102 miRNAs were reported to be upregulated or downregulated. Meta-analysis identified 51 significantly dysregulated miRNAs. The top upregulated miRNA was miR-142-3p (logOR 6.4721; 95% CI 4.9537, 7.9904; adjusted p = 4.60 × 10(-16)). The top downregulated miRNA was miR-126a (logOR 7.5237; 95% CI 4.7159, 10.3316; adjusted p = 3.01 × 10(-07)). The dysregulation of two miRNAs (miR-199a-3p and miR-223) was highly pancreas-specific and liver-specific. miR-30e was downregulated in patients with type 2 diabetes, while miR-92a was downregulated in animal models of diabetes. In sensitivity analysis, 40 out of 47 miRNAs (85%) were robustly and consistently dysregulated. CONCLUSIONS/INTERPRETATION This meta-analysis confirms that 40 miRNAs are significantly dysregulated in type 2 diabetes. miR-29a, miR-34a, miR-375, miR-103, miR-107, miR-132, miR-142-3p and miR-144 are potential circulating biomarkers of type 2 diabetes. In addition, miR-199a-3p and miR-223 are potential tissue biomarkers of type 2 diabetes.
Collapse
Affiliation(s)
- Hongmei Zhu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macao, 999078, China
| | | |
Collapse
|