1
|
Duba M, Al Tukmachi D, Samoilenko T, Vecera M, Ruckova M, Vankova T, Radova L, Kerkovsky M, Dostal M, Koprivova T, Roskova I, Mrlian A, Hrdy O, Duba J, Kren L, Smrcka M, Slaby O, Fadrus P, Sana J. MicroRNA Analysis in Meningiomas with Different Degrees of Tissue Stiffness: A Potential Tool for Effective Preoperative Planning. Neurosurgery 2024:00006123-990000000-01413. [PMID: 39485054 DOI: 10.1227/neu.0000000000003222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 08/26/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Meningioma, the most common primary intracranial tumor, presents challenges in surgical treatment because of varying tissue stiffness. This study explores the molecular background of meningioma stiffness, a critical factor in surgical planning and prognosis, focusing on the utility of microRNAs (miRNAs) as diagnostic biomarkers of tissue stiffness. METHODS Patients with meningiomas treated surgically at the University Hospital Brno were included in this study. Total RNA, isolated from tumor tissue samples, underwent quality control and small RNA sequencing to analyze miRNA expression. Differentially expressed miRNAs were identified, and their association with tumor stiffness was assessed. RESULTS This study identified specific miRNAs differentially expressed in meningiomas with different stiffness levels. Key miRNAs, such as miR-31-5p and miR-34b-5p, showed significant upregulation in stiffer meningiomas. These findings were validated using reverse transcription-quantitative polymerase chain reaction, revealing a potential link between miRNA expression and tumor consistency. The expression of miR-31-5p was most notably associated with the stiffness of the tumor tissue (sensitivity = 71% and specificity = 83%). CONCLUSION This research highlights the potential of miRNAs as biomarkers for determining meningioma tissue stiffness. Identifying specific miRNAs associated with tumor consistency could improve preoperative planning and patient prognosis. These findings pave the way for further exploration of miRNAs in the clinical assessment of meningiomas.
Collapse
Affiliation(s)
- Milos Duba
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Dagmar Al Tukmachi
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Tetiana Samoilenko
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Marek Vecera
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Michaela Ruckova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Tereza Vankova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Lenka Radova
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
| | - Milos Kerkovsky
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Masaryk University, Brno and University Hospital Brno, Brno, Czech Republic
| | - Marek Dostal
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Masaryk University, Brno and University Hospital Brno, Brno, Czech Republic
- Department of Biophysics, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Tereza Koprivova
- Department of Radiology and Nuclear Medicine, Faculty of Medicine, Masaryk University, Brno and University Hospital Brno, Brno, Czech Republic
| | - Ivana Roskova
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Andrej Mrlian
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Ondrej Hrdy
- Department of Anesthesiology, Resuscitation and Intensive Care, University Hospital Brno, Brno, Czech Republic
| | - Jaroslav Duba
- Department of Anesthesiology, Resuscitation and Intensive Care, University Hospital Brno, Brno, Czech Republic
| | - Leos Kren
- Department of Pathology, University Hospital Brno, Brno, Czech Republic
| | - Martin Smrcka
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Pathology, University Hospital Brno, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Pavel Fadrus
- Department of Neurosurgery, University Hospital Brno, Brno, Czech Republic
| | - Jiri Sana
- Central European Institute of Technology, Masaryk University, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Comprehensive Cancer Care, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|
2
|
Abedi S, Behmanesh A, Mazhar FN, Bagherifard A, Sami SH, Heidari N, Hossein-Khannazer N, Namazifard S, Kazem Arki M, Shams R, Zarrabi A, Vosough M. Machine learning and experimental analyses identified miRNA expression models associated with metastatic osteosarcoma. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167357. [PMID: 39033966 DOI: 10.1016/j.bbadis.2024.167357] [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: 04/11/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Osteosarcoma (OS), as the most common primary bone cancer, has a high invasiveness and metastatic potential, therefore, it has a poor prognosis. This study identified early diagnostic biomarkers using miRNA expression profiles associated with osteosarcoma metastasis. In the first step, we used RNA-seq and online microarray data from osteosarcoma tissues and cell lines to identify differentially expressed miRNAs. Then, using seven feature selection algorithms for ranking, the first-ranked miRNAs were selected as input for five machine learning systems. Using network analysis and machine learning algorithms, we developed new diagnostic models that successfully differentiated metastatic osteosarcoma from non-metastatic samples based on newly discovered miRNA signatures. The results showed that miR-34c-3p and miR-154-3p act as the most promising models in the diagnosis of metastatic osteosarcoma. Validation for this model by RT-qPCR in benign tissue and osteosarcoma biopsies confirmed the lower expression of miR-34c-3p and miR-154-3p in OS samples. In addition, a direct correlation between miR-34c-3p expression, miR-154-3p expression and tumor grade was discovered. The combined values of miR-34c-3p and miR-154-3p showed 90 % diagnostic power (AUC = 0.90) for osteosarcoma samples and 85 % (AUC = 0.85) for metastatic osteosarcoma. Adhesion junction and focal adhesion pathways, as well as epithelial-to-mesenchymal transition (EMT) GO terms, were identified as the most significant KEGG and GO terms for the top miRNAs. The findings of this study highlight the potential use of novel miRNA expression signatures for early detection of metastatic osteosarcoma. These findings may help in determining therapeutic approaches with a quantitative and faster method of metastasis detection and also be used in the development of targeted molecular therapy for this aggressive cancer. Further research is needed to confirm the clinical utility of miR-34c-3p and miR-154-3p as diagnostic biomarkers for metastatic osteosarcoma.
Collapse
Affiliation(s)
- Samira Abedi
- Department of Cellular and Molecular Biology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Ali Behmanesh
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Farid Najd Mazhar
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Bagherifard
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Sam Hajialiloo Sami
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Negar Heidari
- Department of Cellular and Molecular Biology, Faculty of Sciences and Advanced Technology in Biology, University of Science and Culture, Tehran, Iran; Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nikoo Hossein-Khannazer
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saina Namazifard
- University of Texas at Arlington, Department of Mechanical and Aerospace Engineering, USA
| | - Mandana Kazem Arki
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roshanak Shams
- Bone and Joint Reconstruction Research Center, Department of Orthopedics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan; Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
3
|
Xu M, Pang M, Wang C, An N, Chen R, Bai Y, He J, Wang C, Qi Y. MiR-92a-3p Knockdown Attenuates Transforming Growth Factor-β1-induced Tubulointerstitial Fibrosis by Targeting LIN28A-mediated EMT Pathway. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:198-206. [PMID: 39148295 DOI: 10.4103/ejpi.ejpi-d-24-00019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/11/2024] [Indexed: 08/17/2024]
Abstract
ABSTRACT The role of microRNAs in regulating tubulointerstitial fibrosis, a key feature of progressive chronic kidney disease, is of significant importance. LIN28A has been reported to attenuate renal fibrosis in obstructive nephropathy. Here, our objective was to investigate the precise biological function of the miR-92a-3p/LIN28A axis in tubulointerstitial fibrosis. The human renal proximal tubular epithelial (HK-2) cell line was exposed to transforming growth factor (TGF)-β1, establishing an in vitro model mimicking tubulointerstitial fibrosis. Luciferase reporter assay was utilized to investigate the relationship between miR-92a-3p and LIN28A. Cell transfection techniques were employed to modify the expression of miR-92a-3p and LIN28A. An in vivo model of tubulointerstitial fibrosis was created by inducing unilateral ureteral obstruction (UUO) in C57BL/6N mice. Our initial observations showed that TGF-β1 treatment of HK-2 cells and the UUO mice model led to an increase in miR-92a-3p expression and a decrease in LIN28A expression. We confirmed that miR-92a-3p directly targeted LIN28A in HK-2 cells. In TGF-β1-stimulated HK-2 cells, knocking down miR-92a-3p notably reduced the levels of alpha smooth muscle actin and vimentin and concurrently enhanced the expression of E-cadherin. These changes were counteracted upon transfection with si-LIN28A. Thus, directing interventions toward miR-92a-3p holds the potential to emerge as a viable therapeutic approach for addressing tubulointerstitial fibrosis.
Collapse
Affiliation(s)
- Mingzhi Xu
- Blood Purification Center, Hainan General Hospital, Hai-nan Affiliated Hospital of Hainan Medical University, Haikou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Garmaa G, Bunduc S, Kói T, Hegyi P, Csupor D, Ganbat D, Dembrovszky F, Meznerics FA, Nasirzadeh A, Barbagallo C, Kökény G. A Systematic Review and Meta-Analysis of microRNA Profiling Studies in Chronic Kidney Diseases. Noncoding RNA 2024; 10:30. [PMID: 38804362 PMCID: PMC11130806 DOI: 10.3390/ncrna10030030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/29/2024] Open
Abstract
Chronic kidney disease (CKD) represents an increasing health burden. Evidence suggests the importance of miRNA in diagnosing CKD, yet the reports are inconsistent. This study aimed to determine novel miRNA biomarkers and potential therapeutic targets from hypothesis-free miRNA profiling studies in human and murine CKDs. Comprehensive literature searches were conducted on five databases. Subgroup analyses of kidney diseases, sample types, disease stages, and species were conducted. A total of 38 human and 12 murine eligible studies were analyzed using Robust Rank Aggregation (RRA) and vote-counting analyses. Gene set enrichment analyses of miRNA signatures in each kidney disease were conducted using DIANA-miRPath v4.0 and MIENTURNET. As a result, top target genes, Gene Ontology terms, the interaction network between miRNA and target genes, and molecular pathways in each kidney disease were identified. According to vote-counting analysis, 145 miRNAs were dysregulated in human kidney diseases, and 32 were dysregulated in murine CKD models. By RRA, miR-26a-5p was significantly reduced in the kidney tissue of Lupus nephritis (LN), while miR-107 was decreased in LN patients' blood samples. In both species, epithelial-mesenchymal transition, Notch, mTOR signaling, apoptosis, G2/M checkpoint, and hypoxia were the most enriched pathways. These miRNA signatures and their target genes must be validated in large patient cohort studies.
Collapse
Affiliation(s)
- Gantsetseg Garmaa
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (G.G.); (A.N.)
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Department of Pathology, School of Medicine, Mongolian National University of Medical Sciences, Ulan-Bator 14210, Mongolia;
| | - Stefania Bunduc
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, Dionisie Lupu Street 37, 020021 Bucharest, Romania
- Fundeni Clinical Institute, Fundeni Street 258, 022328 Bucharest, Romania
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Baross út 22-24, 1085 Budapest, Hungary
| | - Tamás Kói
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Department of Stochastics, Institute of Mathematics, Budapest University of Technology and Economics, Műegyetem rkp. 3, 1111 Budapest, Hungary
| | - Péter Hegyi
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Baross út 22-24, 1085 Budapest, Hungary
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
| | - Dezső Csupor
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Institute for Translational Medicine, Medical School, University of Pécs, 7624 Pécs, Hungary
- Institute of Clinical Pharmacy, University of Szeged, Szikra utca 8, 6725 Szeged, Hungary
| | - Dariimaa Ganbat
- Department of Pathology, School of Medicine, Mongolian National University of Medical Sciences, Ulan-Bator 14210, Mongolia;
- Department of Public Health, Graduate School of Medicine, International University of Health and Welfare, Tokyo 107-840, Japan
| | - Fanni Dembrovszky
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Division of Pancreatic Diseases, Heart and Vascular Center, Semmelweis University, Baross út 22-24, 1085 Budapest, Hungary
| | - Fanni Adél Meznerics
- Center for Translational Medicine, Semmelweis University, Üllői út 26, 1085 Budapest, Hungary; (S.B.); (T.K.); (P.H.); (D.C.); (F.D.); (F.A.M.)
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Mária utca 41, 1085 Budapest, Hungary
| | - Ailar Nasirzadeh
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (G.G.); (A.N.)
| | - Cristina Barbagallo
- Section of Biology and Genetics “G. Sichel”, Department of Biomedical and Biotechnological Sciences, University of Catania, 95123 Catania, Italy;
| | - Gábor Kökény
- Institute of Translational Medicine, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary; (G.G.); (A.N.)
- International Nephrology Research and Training Center, Semmelweis University, Nagyvárad tér 4, 1089 Budapest, Hungary
| |
Collapse
|
5
|
Guiot J, Henket M, Remacle C, Cambier M, Struman I, Winandy M, Moermans C, Louis E, Malaise M, Ribbens C, Louis R, Njock MS. Systematic review of overlapping microRNA patterns in COVID-19 and idiopathic pulmonary fibrosis. Respir Res 2023; 24:112. [PMID: 37061683 PMCID: PMC10105547 DOI: 10.1186/s12931-023-02413-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 04/03/2023] [Indexed: 04/17/2023] Open
Abstract
BACKGROUND Pulmonary fibrosis is an emerging complication of SARS-CoV-2 infection. In this study, we speculate that patients with COVID-19 and idiopathic pulmonary fibrosis (IPF) may share aberrant expressed microRNAs (miRNAs) associated to the progression of lung fibrosis. OBJECTIVE To identify miRNAs presenting similar alteration in COVID-19 and IPF, and describe their impact on fibrogenesis. METHODS A systematic review of the literature published between 2010 and January 2022 (PROSPERO, CRD42022341016) was conducted using the key words (COVID-19 OR SARS-CoV-2) AND (microRNA OR miRNA) or (idiopathic pulmonary fibrosis OR IPF) AND (microRNA OR miRNA) in Title/Abstract. RESULTS Of the 1988 references considered, 70 original articles were appropriate for data extraction: 27 studies focused on miRNAs in COVID-19, and 43 on miRNAs in IPF. 34 miRNAs were overlapping in COVID-19 and IPF, 7 miRNAs presenting an upregulation (miR-19a-3p, miR-200c-3p, miR-21-5p, miR-145-5p, miR-199a-5p, miR-23b and miR-424) and 9 miRNAs a downregulation (miR-17-5p, miR-20a-5p, miR-92a-3p, miR-141-3p, miR-16-5p, miR-142-5p, miR-486-5p, miR-708-3p and miR-150-5p). CONCLUSION Several studies reported elevated levels of profibrotic miRNAs in COVID-19 context. In addition, the balance of antifibrotic miRNAs responsible of the modulation of fibrotic processes is impaired in COVID-19. This evidence suggests that the deregulation of fibrotic-related miRNAs participates in the development of fibrotic lesions in the lung of post-COVID-19 patients.
Collapse
Affiliation(s)
- Julien Guiot
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Fibropole Research Group, University Hospital of Liège, Liège, Belgium
| | - Monique Henket
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Claire Remacle
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Laboratory of Molecular Angiogenesis, GIGA Research Center, University of Liège, Liège, Belgium
| | - Maureen Cambier
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Laboratory of Molecular Angiogenesis, GIGA Research Center, University of Liège, Liège, Belgium
| | - Ingrid Struman
- Laboratory of Molecular Angiogenesis, GIGA Research Center, University of Liège, Liège, Belgium
| | - Marie Winandy
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Catherine Moermans
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
| | - Edouard Louis
- Laboratory of Gastroenterology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Fibropole Research Group, University Hospital of Liège, Liège, Belgium
| | - Michel Malaise
- Laboratory of Rheumatology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Fibropole Research Group, University Hospital of Liège, Liège, Belgium
| | - Clio Ribbens
- Laboratory of Rheumatology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Fibropole Research Group, University Hospital of Liège, Liège, Belgium
| | - Renaud Louis
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Fibropole Research Group, University Hospital of Liège, Liège, Belgium
| | - Makon-Sébastien Njock
- Laboratory of Pneumology, GIGA Research Center, University of Liège, University Hospital of Liège, Liège, Belgium
- Fibropole Research Group, University Hospital of Liège, Liège, Belgium
| |
Collapse
|
6
|
Saito S, Ohno SI, Harada Y, Kanno Y, Kuroda M. MiR-34a induces myofibroblast differentiation from renal fibroblasts. Clin Exp Nephrol 2023; 27:411-418. [PMID: 36808381 DOI: 10.1007/s10157-023-02329-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/07/2023] [Indexed: 02/21/2023]
Abstract
BACKGROUND Renal fibrosis is the common outcome of progressive kidney diseases. To avoid dialysis, the molecular mechanism of renal fibrosis must be explored further. MicroRNAs play key roles in renal fibrosis. MiR-34a is a transcriptional target of p53, which regulates the cell cycle and apoptosis. Previous studies demonstrated that miR-34a promotes renal fibrosis. However, the distinct roles of miR-34a in renal fibrosis have not been fully elucidated. Here, we identified the roles of miR-34a in renal fibrosis. METHOD We first analyzed p53 and miR-34a expression in kidney tissues in s UUO (unilateral ureteral obstruction) mouse model. Then, to confirm the effects of miR-34a in vitro, we transfected a miR-34a mimic into a kidney fibroblast cell line (NRK-49F) and analyzed. RESULTS We found that the expression of p53 and miR-34a was upregulated after UUO. Furthermore, after transfection of the miR-34a mimic into kidney fibroblasts, the expression of α-SMA was upregulated dramatically. In addition, α-SMA upregulation was greater upon transfection of the miR-34a mimic than upon treatment with TGF-β1. Moreover, high expression of Acta2 was maintained despite sufficient removal of the miR-34a mimic by changing the medium 4 times during the 9-day culture. After transfection of the miR-34a mimic into kidney fibroblasts, we did not detect phospho-SMAD2/3 by immunoblotting analysis. CONCLUSION Our study revealed that miR-34a induces myofibroblast differentiation from renal fibroblasts. Moreover, the miR-34a-induced upregulation of α-SMA was independent of the TGF-β/SMAD signaling pathway. In conclusion, our study indicated that the p53/miR-34a axis promotes the development of renal fibrosis.
Collapse
Affiliation(s)
- Suguru Saito
- Department of Nephrology, Tokyo Medical University, Tokyo, Japan
| | - Shin-Ichiro Ohno
- Deparatment of Molecular Pathology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan.
| | - Yuichirou Harada
- Deparatment of Molecular Pathology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| | - Yoshihiko Kanno
- Department of Nephrology, Tokyo Medical University, Tokyo, Japan
| | - Masahiko Kuroda
- Deparatment of Molecular Pathology, Tokyo Medical University, 6-1-1, Shinjuku, Shinjuku-ku, Tokyo, 160-8402, Japan
| |
Collapse
|
7
|
Kaneko S, Yanai K, Ishii H, Aomatsu A, Hirai K, Ookawara S, Ishibashi K, Morishita Y. miR-122-5p Regulates Renal Fibrosis In Vivo. Int J Mol Sci 2022; 23:ijms232315423. [PMID: 36499744 PMCID: PMC9736395 DOI: 10.3390/ijms232315423] [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: 11/19/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/12/2022] Open
Abstract
The role of exogenous microRNAs (miRNAs) in renal fibrosis is poorly understood. Here, the effect of exogenous miRNAs on renal fibrosis was investigated using a renal fibrosis mouse model generated by unilateral ureteral obstruction (UUO). miRNA microarray analysis and quantitative reverse-transcription polymerase chain reaction showed that miR-122-5p was the most downregulated (0.28-fold) miRNA in the kidneys of UUO mice. The injection of an miR-122-5p mimic promoted renal fibrosis and upregulated COL1A2 and FN1, whereas an miR-122-5p inhibitor suppressed renal fibrosis and downregulated COL1A2 and FN1. The expression levels of fibrosis-related mRNAs, which were predicted targets of miR-122-5p, were evaluated. The expression level of TGFBR2, a pro-fibrotic mRNA, was upregulated by the miR-122-5p mimic, and the expression level of FOXO3, an anti-fibrotic mRNA, was upregulated by the miR-122-5p inhibitor. The protein expressions of TGFBR2 and FOXO3 were confirmed by immunohistochemistry. Additionally, the expression levels of LC3, downstream anti-fibrotic mRNAs of FOXO3, were upregulated by the miR-122-5p inhibitor. These results suggest that miR-122-5p has critical roles in renal fibrosis.
Collapse
Affiliation(s)
- Shohei Kaneko
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Katsunori Yanai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Hiroki Ishii
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Akinori Aomatsu
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
- Division of Intensive Care Unit, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Keiji Hirai
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Susumu Ookawara
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
| | - Kenichi Ishibashi
- Department of Medical Physiology, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Yoshiyuki Morishita
- Division of Nephrology, First Department of Integrated Medicine, Saitama Medical Center, Jichi Medical University, Saitama 330-8503, Japan
- Correspondence:
| |
Collapse
|
8
|
Gupta N, Matsumoto T, Hiratsuka K, Saiz EG, Zhang C, Galichon P, Miyoshi T, Susa K, Tatsumoto N, Yamashita M, Morizane R. Modeling injury and repair in kidney organoids reveals that homologous recombination governs tubular intrinsic repair. Sci Transl Med 2022; 14:eabj4772. [PMID: 35235339 PMCID: PMC9161367 DOI: 10.1126/scitranslmed.abj4772] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Kidneys have the capacity for intrinsic repair, preserving kidney architecture with return to a basal state after tubular injury. When injury is overwhelming or repetitive, however, that capacity is exceeded and incomplete repair results in fibrotic tissue replacing normal kidney parenchyma. Loss of nephrons correlates with reduced kidney function, which defines chronic kidney disease (CKD) and confers substantial morbidity and mortality to the worldwide population. Despite the identification of pathways involved in intrinsic repair, limited treatments for CKD exist, partly because of the limited throughput and predictivity of animal studies. Here, we showed that kidney organoids can model the transition from intrinsic to incomplete repair. Single-nuclear RNA sequencing of kidney organoids after cisplatin exposure identified 159 differentially expressed genes and 29 signal pathways in tubular cells undergoing intrinsic repair. Homology-directed repair (HDR) genes including Fanconi anemia complementation group D2 (FANCD2) and RAD51 recombinase (RAD51) were transiently up-regulated during intrinsic repair but were down-regulated in incomplete repair. Single cellular transcriptomics in mouse models of obstructive and hemodynamic kidney injury and human kidney samples of immune-mediated injury validated HDR gene up-regulation during tubular repair. Kidney biopsy samples with tubular injury and varying degrees of fibrosis confirmed loss of FANCD2 during incomplete repair. Last, we performed targeted drug screening that identified the DNA ligase IV inhibitor, SCR7, as a therapeutic candidate that rescued FANCD2/RAD51-mediated repair to prevent the progression of CKD in the cisplatin-induced organoid injury model. Our findings demonstrate the translational utility of kidney organoids to identify pathologic pathways and potential therapies.
Collapse
Affiliation(s)
- Navin Gupta
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, USA
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Takuya Matsumoto
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Ken Hiratsuka
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Edgar Garcia Saiz
- Harvard Medical School, Boston, MA, USA
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Chengcheng Zhang
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Pierre Galichon
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, USA
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Tomoya Miyoshi
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Koichiro Susa
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| | - Narihito Tatsumoto
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Michifumi Yamashita
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Ryuji Morizane
- Nephrology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Harvard Stem Cell Institute (HSCI), Cambridge, MA, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA
- Nephrology Division, Department of Medicine, Brigham and Women’s Hospital, Boston, MA, USA
| |
Collapse
|
9
|
Hanusek K, Rybicka B, Popławski P, Adamiok-Ostrowska A, Głuchowska K, Piekiełko-Witkowska A, Bogusławska J. TGF‑β1 affects the renal cancer miRNome and regulates tumor cells proliferation. Int J Mol Med 2022; 49:52. [PMID: 35179216 PMCID: PMC8904080 DOI: 10.3892/ijmm.2022.5108] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/19/2022] [Indexed: 11/24/2022] Open
Abstract
TGF-β1 is a pleiotropic cytokine that can either promote or inhibit cancer development and progression. It was previously found that TGF-β1 can regulate the expression of several microRNAs (miR or miRNA) involved in the progression of renal cell carcinoma (RCC). Therefore, the present study aimed to analyze the effects of TGF-β1 on the global RCC miRNome. It was found that TGF-β1 can regulate a complex network consisting of miRNAs and mRNAs involved in RCC transformation. In particular, TGF-β1 was revealed to regulate the proliferation of RCC cells while concomitantly modifying the expression of oncogenic regulators, including avian erythroblastosis virus E26 (V-Ets) oncogene homolog-1 (ETS1). In addition, TGF-β1 was demonstrated to regulate the expression of a number of miRNAs including miR-30c-5p, miR-155-5p, miR-181a-5p and miR-181b-5p. By contrast, TGF-β1 reciprocally modified the expression of genes encoding TGF-β1 receptors and SMADs, indicating a novel regulatory feedback mechanism mediated through the miRNAs. These data suggested that ETS1 served different roles in different subtypes of RCC tumors, specifically by functioning as an oncogene in clear cell RCC while as a tumor suppressor in papillary RCC.
Collapse
Affiliation(s)
- Karolina Hanusek
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01‑813 Warsaw, Poland
| | - Beata Rybicka
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01‑813 Warsaw, Poland
| | - Piotr Popławski
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01‑813 Warsaw, Poland
| | - Anna Adamiok-Ostrowska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01‑813 Warsaw, Poland
| | - Katarzyna Głuchowska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01‑813 Warsaw, Poland
| | | | - Joanna Bogusławska
- Department of Biochemistry and Molecular Biology, Centre of Postgraduate Medical Education, 01‑813 Warsaw, Poland
| |
Collapse
|
10
|
Hu X, Shen N, Liu A, Wang W, Zhang L, Sui Z, Tang Q, Du X, Yang N, Ying W, Qin B, Li Z, Li L, Wang N, Lin H. Bone marrow mesenchymal stem cell-derived exosomal miR-34c-5p ameliorates RIF by inhibiting the core fucosylation of multiple proteins. Mol Ther 2022; 30:763-781. [PMID: 34678513 PMCID: PMC8821970 DOI: 10.1016/j.ymthe.2021.10.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/02/2021] [Accepted: 10/10/2021] [Indexed: 02/07/2023] Open
Abstract
Renal interstitial fibrosis (RIF) is an incurable pathological lesion in chronic kidney diseases. Pericyte activation is the major pathological characteristic of RIF. Fibroblast and macrophage activation are also involved in RIF. Studies have revealed that core fucosylation (CF), an important post-translational modification of proteins, plays a key role in pericyte activation and RIF by regulating multiple profibrotic signaling pathways as a hub-like target. Here, we reveal that mesenchymal stem cell (MSC)-derived exosomes reside specifically in the injured kidney and deliver microRNA (miR)-34c-5p to reduce cellular activation and RIF by inhibiting CF. Furthermore, we showed that the CD81-epidermal growth factor receptor (EGFR) ligand-receptor complex aids the entry of exosomal miR-34c-5p into pericytes, fibroblasts, and macrophages. Altogether, our findings reveal a novel role of MSC-derived exosomes in inhibiting multicellular activation via CF and provide a potential intervention strategy for renal fibrosis.
Collapse
Affiliation(s)
- Xuemei Hu
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China,Graduate School of Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian 116044, China
| | - Nan Shen
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China
| | - Anqi Liu
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China,Graduate School of Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian 116044, China
| | - Weidong Wang
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China
| | - Lihua Zhang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Zhigang Sui
- CAS Key Laboratory of Separation Science for Analytical Chemistry, National Chromatographic R&A Center, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Qingzhu Tang
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China
| | - Xiangning Du
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China
| | - Ning Yang
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China
| | - Wantao Ying
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 102206 China
| | - Biaojie Qin
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China
| | - Zhitong Li
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China,Graduate School of Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian 116044, China
| | - Lin Li
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China,Graduate School of Dalian Medical University, 9 Western Section, Lvshun South Street, Lvshunkou District, Dalian 116044, China
| | - Nan Wang
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China,Corresponding author: Nan Wang, Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian, 116011, China.
| | - Hongli Lin
- Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian 116011, China,Corresponding author: Hongli Lin, Department of Nephrology, The First Affiliated Hospital of Dalian Medical University, Key Laboratory of Kidney Disease of Liaoning Province, The Center for the Transformation Medicine of Kidney Disease of Liaoning Province, No. 222 Zhongshan Road, Dalian, 116011, China.
| |
Collapse
|
11
|
Abstract
Epigenetics examines heritable changes in DNA and its associated proteins except mutations in gene sequence. Epigenetic regulation plays fundamental roles in kidney cell biology through the action of DNA methylation, chromatin modification via epigenetic regulators and non-coding RNA species. Kidney diseases, including acute kidney injury, chronic kidney disease, diabetic kidney disease and renal fibrosis are multistep processes associated with numerous molecular alterations even in individual kidney cells. Epigenetic alterations, including anomalous DNA methylation, aberrant histone alterations and changes of microRNA expression all contribute to kidney pathogenesis. These changes alter the genome-wide epigenetic signatures and disrupt essential pathways that protect renal cells from uncontrolled growth, apoptosis and development of other renal associated syndromes. Molecular changes impact cellular function within kidney cells and its microenvironment to drive and maintain disease phenotype. In this chapter, we briefly summarize epigenetic mechanisms in four kidney diseases including acute kidney injury, chronic kidney disease, diabetic kidney disease and renal fibrosis. We primarily focus on current knowledge about the genome-wide profiling of DNA methylation and histone modification, and epigenetic regulation on specific gene(s) in the pathophysiology of these diseases and the translational potential of identifying new biomarkers and treatment for prevention and therapy. Incorporating epigenomic testing into clinical research is essential to elucidate novel epigenetic biomarkers and develop precision medicine using emerging therapies.
Collapse
|
12
|
Chavda V, Chaurasia B, Deora H, Umana GE. Chronic Kidney disease and stroke: A Bi-directional risk cascade and therapeutic update. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
13
|
Negative regulators of TGF-β1 signaling in renal fibrosis; pathological mechanisms and novel therapeutic opportunities. Clin Sci (Lond) 2021; 135:275-303. [PMID: 33480423 DOI: 10.1042/cs20201213] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/23/2020] [Accepted: 01/08/2021] [Indexed: 02/06/2023]
Abstract
Elevated expression of the multifunctional cytokine transforming growth factor β1 (TGF-β1) is causatively linked to kidney fibrosis progression initiated by diabetic, hypertensive, obstructive, ischemic and toxin-induced injury. Therapeutically relevant approaches to directly target the TGF-β1 pathway (e.g., neutralizing antibodies against TGF-β1), however, remain elusive in humans. TGF-β1 signaling is subjected to extensive negative control at the level of TGF-β1 receptor, SMAD2/3 activation, complex assembly and promoter engagement due to its critical role in tissue homeostasis and numerous pathologies. Progressive kidney injury is accompanied by the deregulation (loss or gain of expression) of several negative regulators of the TGF-β1 signaling cascade by mechanisms involving protein and mRNA stability or epigenetic silencing, further amplifying TGF-β1/SMAD3 signaling and fibrosis. Expression of bone morphogenetic proteins 6 and 7 (BMP6/7), SMAD7, Sloan-Kettering Institute proto-oncogene (Ski) and Ski-related novel gene (SnoN), phosphate tensin homolog on chromosome 10 (PTEN), protein phosphatase magnesium/manganese dependent 1A (PPM1A) and Klotho are dramatically decreased in various nephropathies in animals and humans albeit with different kinetics while the expression of Smurf1/2 E3 ligases are increased. Such deregulations frequently initiate maladaptive renal repair including renal epithelial cell dedifferentiation and growth arrest, fibrotic factor (connective tissue growth factor (CTGF/CCN2), plasminogen activator inhibitor type-1 (PAI-1), TGF-β1) synthesis/secretion, fibroproliferative responses and inflammation. This review addresses how loss of these negative regulators of TGF-β1 pathway exacerbates renal lesion formation and discusses the therapeutic value in restoring the expression of these molecules in ameliorating fibrosis, thus, presenting novel approaches to suppress TGF-β1 hyperactivation during chronic kidney disease (CKD) progression.
Collapse
|
14
|
Sargazi S, Heidari Nia M, Mirani Sargazi F, Sheervalilou R, Saravani R, Bahrami S, Mirinejad S, Alidadi A. Functional miR143/145 Cluster Variants and Haplotypes Are Associated with Chronic Kidney Disease: a Preliminary Case-Control Study and Computational Analyses. Appl Biochem Biotechnol 2021; 193:1532-1544. [PMID: 33484447 DOI: 10.1007/s12010-021-03489-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/07/2021] [Indexed: 12/19/2022]
Abstract
MiR-143/145 cluster is a novel transcriptional target of many signaling pathways, with variations within this cluster contributed to the risk of multiple diseases. To date, no data regarding the link between miR143/145 cluster polymorphisms and the risk of developing chronic kidney disease (CKD) has been reported. Hence, we aimed to examine such association in a population of Iranian ancestry. In this preliminary study, 276 CKD patients and 300 unrelated age and sex-matched healthy controls were recruited. Genotyping was performed by PCR-RFLP and allele-specific-PCR methods. Computational analyses were performed to predict the potential effects of the variants. Our findings indicated that rs41291957, rs12659504, and rs353292 polymorphisms were positively associated with CKD, while rs4705342 and rs4705343 polymorphisms demonstrated a significant negative association with the disease. Moreover, a significant association was observed between CC + TC and TT genotypes and CKD stages. We found that AACTT, AATTC, AATTT, GATTC, GATTT, and GGCTT haplotypes significantly enhanced the risk of CKD compared with the Grs41291957AArs12659504Crs353292Trs4705342Trs4705343 haplotype. Computational analysis showed that rs353292, rs4705342, and rs4705343 might alter the binding of the transcription factors in this gene cluster. We found that miR-143/145 cluster polymorphisms were associated with CKD risk in a sample of the Iranian population. Replicated studies on different ethnicities are necessary to investigate the association between these promoter variants and clinical outcomes.
Collapse
Affiliation(s)
- Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Milad Heidari Nia
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Fariba Mirani Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Roghayeh Sheervalilou
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran.
- Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Sara Bahrami
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ali Alidadi
- Nephrology Department, Faculty of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| |
Collapse
|
15
|
Bai L, Lin Y, Xie J, Zhang Y, Wang H, Zheng D. MiR-27b-3p inhibits the progression of renal fibrosis via suppressing STAT1. Hum Cell 2021; 34:383-393. [PMID: 33454903 PMCID: PMC7900087 DOI: 10.1007/s13577-020-00474-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 12/14/2020] [Indexed: 01/02/2023]
Abstract
Renal fibrosis is a pathologic change in chronic kidney disease (CKD). MicroRNAs (miRNAs) have been shown to play an important role in the development of renal fibrosis. However, the biological role of miR-27b-3p in renal fibrosis remains unclear. Thus, this study aimed to investigate the role of miR-27b-3p in the progression of renal fibrosis. In this study, HK-2 cells were stimulated with transforming growth factor (TGF)-β1 for mimicking fibrosis progression in vitro. The unilateral ureteric obstruction (UUO)-induced mice renal fibrosis in vivo was established as well. The results indicated that the overexpression of miR-27b-3p significantly inhibited epithelial-to-mesenchymal transition (EMT) in TGF-β1-stimulated HK-2 cells, as shown by the decreased expressions of α-SMA, collagen III, Fibronectin and Vimentin. In addition, overexpression of miR-27b-3p markedly decreased TGF-β1-induced apoptosis in HK-2 cells, as evidenced by the decreased levels of Fas, active caspase 8 and active caspase 3. Meanwhile, dual-luciferase assay showed that miR-27b-3p downregulated signal transducers and activators of transcription 1 (STAT1) expression through direct binding with the 3′-UTR of STAT1. Furthermore, overexpression of miR-27b-3p attenuated UUO-induced renal fibrosis via downregulation of STAT1, α-SMA and collagen III. In conclusion, miR-27b-3p overexpression could alleviate renal fibrosis via suppressing STAT1 in vivo and in vitro. Therefore, miR-27b-3p might be a promising therapeutic target for the treatment of renal fibrosis.
Collapse
Affiliation(s)
- Lin Bai
- Department of Nephrology, Affiliated Huai'an Hospital of Xuzhou Medical University, 62# Huaihai South Road, Huai'an, 223001, Jiangsu, People's Republic of China
| | - Yongtao Lin
- Department of Nephrology, Affiliated Huai'an Hospital of Xuzhou Medical University, 62# Huaihai South Road, Huai'an, 223001, Jiangsu, People's Republic of China
| | - Juan Xie
- Department of Nephrology, Affiliated Huai'an Hospital of Xuzhou Medical University, 62# Huaihai South Road, Huai'an, 223001, Jiangsu, People's Republic of China
| | - Yiyuan Zhang
- Xuzhou Medical University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Hongwu Wang
- Department of Nephrology, Affiliated Huai'an Hospital of Xuzhou Medical University, 62# Huaihai South Road, Huai'an, 223001, Jiangsu, People's Republic of China.
| | - Donghui Zheng
- Department of Nephrology, Affiliated Huai'an Hospital of Xuzhou Medical University, 62# Huaihai South Road, Huai'an, 223001, Jiangsu, People's Republic of China.
| |
Collapse
|
16
|
Wu T, Xie D, Zhao X, Xu M, Luo L, Deng D, Chen M. Enhanced Expression of miR-34c in Peripheral Plasma Associated with Diabetic Foot Ulcer in Type 2 Diabetes Patients. Diabetes Metab Syndr Obes 2021; 14:4263-4273. [PMID: 34703259 PMCID: PMC8526515 DOI: 10.2147/dmso.s326066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 08/25/2021] [Indexed: 04/20/2023] Open
Abstract
OBJECTIVE To explore the correlation between the expression of miR-34c in peripheral blood of patients with type 2 diabetes mellitus (T2DM) and the onset of diabetic foot ulcer (DFU) and diabetic foot osteomyelitis (DFO). METHODS Sixty newly diagnosed patients with T2DM without DFU (T2DM group), 112 T2DM patients with DFU (DFU group) and 60 controls with normal glucose tolerance (NC group). The DFU group patients were subdivided into DFO (n=64) and NDFO (n=48) groups. Quantitative real-time PCR (qRT-PCR) method was used to determine miR-34c expression levels in the peripheral blood of subjects to analyze the clinical characteristics of DFU and DFO risk factors. RESULTS MiR-34c expression level in the T2DM group was marked higher than the NC group [2.99 (1.45-6.22) vs 1.01 (0.89-1.52)] (P < 0.05). However, the expression level of miR-34c in the DFU group was significantly higher than the T2DM group [9.65 (6.15-18.63) vs 2.99 (1.45-6.22)] (P < 0.01). Compared with the NDFO group, the expression level of miR-34c in the DFO group was also obviously increased [13.46 (8.89-19.11) vs 6.02 (5.93-14.72)] (P < 0.01). The expression level of miR-34c in DFU patients was positively correlated with the amputation rate of foot ulcers (P=0.030) and was negatively correlated with the healing rate of foot ulcers after eight weeks (P=0.025). Multifactorial logistic regression analysis showed that increased expression of miR-34c was an independent risk factor for DFU and DFO (ORDFU=3.47, ORDFO=4.25, P < 0.01). Meanwhile, the ROC curve analysis indicated that the AUC of miR-34c for the diagnosis of DFU and DFO was 0.803 and 0.904, the optimum sensitivity being was 100% and 98.7%, the optimum specificity was 98.4% and 98.4%, respectively. CONCLUSION The increased expression of miR-34c in peripheral blood of T2DM patients is closely related to the occurrence, development and prognosis of DFU and DFO.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Dandan Xie
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Xiaotong Zhao
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Murong Xu
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Li Luo
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Datong Deng
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
| | - Mingwei Chen
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230032, People’s Republic of China
- Correspondence: Mingwei Chen Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, No. 218 Jixi Road, Hefei, Anhui, 230032, People’s Republic of China Email
| |
Collapse
|
17
|
Zhang B, Zhao C, Hou L, Wu Y. Silencing of the lncRNA TUG1 attenuates the epithelial-mesenchymal transition of renal tubular epithelial cells by sponging miR-141-3p via regulating β-catenin. Am J Physiol Renal Physiol 2020; 319:F1125-F1134. [PMID: 33135476 DOI: 10.1152/ajprenal.00321.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Renal interstitial fibrosis (RIF) is characterized by excessive extracellular matrix deposition and involves epithelial-mesenchymal transition (EMT). The lncRNA taurine-upregulated gene 1 (TUG1) participates in EMT in several cancers; however, the effect and underlying mechanism of TUG1 in RIF-related EMT remain unclear. Here, we explored the mechanisms by which TUG1 modulates RIF. An in vivo model of renal fibrosis was established by unilateral ureteral obstruction in Balb/c mice. Human renal proximal tubular epithelial (HK-2) cells treated with transforming growth factor (TGF)-β1 were used to induce the in vitro model. Morphological changes and TUG1 expression were assessed. HK-2 cells were transfected with siRNA to silence TUG1. Western blot analysis, immunofluorescence staining, cell proliferation, and migration assays were performed to examine TGF-β1-induced changes in EMT markers and EMT-like cell behaviors. TUG1 and β-catenin (CTNNB1) levels were significantly upregulated, whereas miR-141-3p was significantly downregulated, during EMT in vitro and in vivo. TUG1 knockdown or miR-141-3p overexpression supported the epithelioid morphology of HK-2 cells while enhancing the downregulation of E-cadherin and upregulation of vimentin, α-smooth muscle actin, and β-catenin levels in TGF-β1-treated HK-2 cells. TUG1 knockdown promoted the proliferation and decreased the migration of HK-2 cells and enhanced the downregulation of miR-141-3p levels in TGF-β1-treated HK-2 cells. TUG1 directly targeted miR-141-3p, and miR-141-3p was directly bound to CTNNB1. Downregulation of miR-141-3p inhibited TUG1 silencing-induced suppression of EMT. In conclusion, TUG1 promotes EMT in TGF-β1-induced HK-2 cells via upregulation of β-catenin levels by sponging miR-141-3p, suggesting a novel therapeutic candidate for RIF.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chengguang Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ling Hou
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yubin Wu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Carter LE, Cook DP, Collins O, Gamwell LF, Dempster HA, Wong HW, McCloskey CW, Garson K, Vuong NH, Vanderhyden BC. COX2 is induced in the ovarian epithelium during ovulatory wound repair and promotes cell survival†. Biol Reprod 2020; 101:961-974. [PMID: 31347667 DOI: 10.1093/biolre/ioz134] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 07/02/2019] [Accepted: 07/17/2019] [Indexed: 12/18/2022] Open
Abstract
The ovarian surface epithelium (OSE) is a monolayer of cells surrounding the ovary that is ruptured during ovulation. After ovulation, the wound is repaired, however, this process is poorly understood. In epithelial tissues, wound repair is mediated by an epithelial-to-mesenchymal transition (EMT). Transforming Growth Factor Beta-1 (TGFβ1) is a cytokine commonly known to induce an EMT and is present throughout the ovarian microenvironment. We, therefore, hypothesized that TGFβ1 induces an EMT in OSE cells and activates signaling pathways important for wound repair. Treating primary cultures of mouse OSE cells with TGFβ1 induced an EMT mediated by TGFβRI signaling. The transcription factor Snail was the only EMT-associated transcription factor increased by TGFβ1 and, when overexpressed, was shown to increase OSE cell migration. A polymerase chain reaction array of TGFβ signaling targets determined Cyclooxygenase-2 (Cox2) to be most highly induced by TGFβ1. Constitutive Cox2 expression modestly increased migration and robustly enhanced cell survival, under stress conditions similar to those observed during wound repair. The increase in Snail and Cox2 expression with TGFβ1 was reproduced in human OSE cultures, suggesting these responses are conserved between mouse and human. Finally, the induction of Cox2 expression in OSE cells during ovulatory wound repair was shown in vivo, suggesting TGFβ1 increases Cox2 to promote wound repair by enhancing cell survival. These data support that TGFβ1 promotes ovulatory wound repair by induction of an EMT and activation of a COX2-mediated pro-survival pathway. Understanding ovulatory wound repair may give insight into why ovulation is the primary non-hereditary risk factor for ovarian cancer.
Collapse
Affiliation(s)
- Lauren E Carter
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David P Cook
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Olga Collins
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Lisa F Gamwell
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Holly A Dempster
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Howard W Wong
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Curtis W McCloskey
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Ken Garson
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Nhung H Vuong
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Barbara C Vanderhyden
- Cancer Therapeutics Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Department of Obstetrics and Gynecology, University of Ottawa/The Ottawa Hospital, Ottawa, Ontario, Canada
| |
Collapse
|
19
|
Down-regulation of LINC00667 hinders renal tubular epithelial cell apoptosis and fibrosis through miR-34c. Clin Transl Oncol 2020; 23:572-581. [PMID: 32705492 DOI: 10.1007/s12094-020-02451-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 07/03/2020] [Indexed: 01/11/2023]
Abstract
PURPOSE This study aimed to down-regulate LINC00667 and inhibit apoptosis and fibrosis of renal tubular epithelial cells through miR-34c. METHODS Altogether, 98 patients with chronic kidney disease treated in our hospital were selected as the study group, and 67 normal people were selected as the control group. Epithelial cells of proximal convoluted tubules in human renal cortex were purchased. TGF-β1 was used to induce fibrosis of HK-2 renal tubular epithelial cells. The expression of LINC00667, miR-34c, type I collagen (Col 1) and type III collagen (Col 3) were detected by qRT-PCR and WB. RESULTS LINC00667 was highly expressed in cancer tissues and HK-2, while miR-34c was poorly expressed. Inhibition of LINC00667 and over-expression of miR-34c could inhibit the proliferation and invasion of chronic kidney disease cells, but increase the apoptosis rate. Down-regulation of LINC00667 could significantly reduce of Col 1 and Col 3 in renal interstitial fibroblasts induced by TGF-β1, while up-regulation of miR-34c could also achieve this effect. Double luciferase report confirmed that there was a targeted regulatory relationship between LINC00667 and miR-34c. CONCLUSION LINC00667 could reduce the proliferation and invasion of chronic kidney disease cells, increase the apoptosis rate by regulating miR-34c, and improve renal fibrosis.
Collapse
|
20
|
Rayego-Mateos S, Valdivielso JM. New therapeutic targets in chronic kidney disease progression and renal fibrosis. Expert Opin Ther Targets 2020; 24:655-670. [PMID: 32338087 DOI: 10.1080/14728222.2020.1762173] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
INTRODUCTION The current therapeutic armamentarium to prevent chronic kidney disease (CKD) progression is limited to the control of blood pressure and in diabetic patients, the strict control of glucose levels. Current research is primarily focused on the reduction of inflammation and fibrosis at different levels. AREAS COVERED This article examines the latest progress in this field and places an emphasis on inflammation, oxidative stress, and fibrosis. New therapeutic targets are described and evidence from experimental and clinical studies is summarized. We performed a search in Medline for articles published over the last 10 years. EXPERT OPINION The search for therapeutic targets of renal inflammation is hindered by an incomplete understanding of the pathophysiology. The determination of the specific inducers of inflammation in the kidney is an area of heightened potential. Prevention of the progression of renal fibrosis by blocking TGF-β signaling has been unsuccessful, but the investigation of signaling pathways involved in late stages of fibrosis progression could yield improved results. Preventive strategies such as the modification of microbiota-inducers of uremic toxins involved in CKD progression is a promising field because of the interaction between the gut microbiota and the renal system.
Collapse
Affiliation(s)
- Sandra Rayego-Mateos
- Red De Investigación Renal (Redinren) , Spain.,Vascular and Renal Translational Research Group, Institut De Recerca Biomèdica De Lleida IRBLleida , Lleida, Spain
| | - Jose M Valdivielso
- Red De Investigación Renal (Redinren) , Spain.,Vascular and Renal Translational Research Group, Institut De Recerca Biomèdica De Lleida IRBLleida , Lleida, Spain
| |
Collapse
|
21
|
Waterborg CEJ, Broeren MGA, Blaney Davidson EN, Koenders MI, van Lent PLEM, van den Berg WB, van der Kraan PM, van de Loo FAJ. The level of synovial AXL expression determines the outcome of inflammatory arthritis, possibly depending on the upstream role of TGF-β1. Rheumatology (Oxford) 2020; 58:536-546. [PMID: 30508140 DOI: 10.1093/rheumatology/key337] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 10/15/2018] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE To investigate the role of AXL, a member of the anti-inflammatory TYRO3, AXL MER (TAM) receptor family, in arthritis. METHODS KRN serum transfer arthritis was induced in Axl-/- and wild-type mice. Knee and ankle joints were scored macro- and microscopically. Synovial gene and protein expression of Axl was determined in naïve and TGF-β1-overexpressing joints. AXL expression was determined in M1-like or M2-like macrophages and RA synovium. Human macrophages, fibroblasts and synovial micromasses were stimulated with TGF-β1 or the AXL inhibitor R428. RESULTS Ankle joints of Axl-/- mice showed exacerbated arthritis pathology, whereas no effect of Axl gene deletion was observed on gonarthritis pathology. To explain this spatial difference, we examined the synovium of naïve mice. In contrast to the knee, the ankle synovial cells prominently expressed AXL. Moreover, the M2-like macrophage phenotype was the dominant cell type in the naïve ankle joint. Human M2-like macrophages expressed higher levels of AXL and blocking AXL increased their inflammatory response. In the murine ankle synovium, gene expression of Tgfb1 was increased and Tgb1 correlated with Axl. Moreover, TGFB1 and AXL expression also correlated in human RA synovium. In human macrophages and synovial micromasses, TGF-β1 enhanced AXL expression. Moreover, TGF-β1 overexpression in naïve murine knee joints induced synovial AXL expression. CONCLUSION Differences in synovial AXL expression are in accordance with the observation that AXL dampens arthritis in ankle, but not in knee joints. We provide evidence that the local differences in AXL expression could be due to TGF-β1, and suggest similar pathways operate in RA synovium.
Collapse
Affiliation(s)
- Claire E J Waterborg
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Mathijs G A Broeren
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Esmeralda N Blaney Davidson
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Marije I Koenders
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter L E M van Lent
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Wim B van den Berg
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Peter M van der Kraan
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fons A J van de Loo
- Experimental Rheumatology, Department of Rheumatology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
22
|
Mu Y, Zhang J, Liu Y, Ma J, Jiang D, Zhang X, Yi X, Cheng K, Shen S, Yang Y, Zhuang R, Zhang Y. CD226 deficiency on regulatory T cells aggravates renal fibrosis via up‐regulation of Th2 cytokines through miR‐340. J Leukoc Biol 2019; 107:573-587. [PMID: 31802539 DOI: 10.1002/jlb.2ma1119-174rr] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/21/2019] [Accepted: 11/22/2019] [Indexed: 12/11/2022] Open
Affiliation(s)
- Yang Mu
- Department of Immunology Fourth Military Medical University Xi'an Shaanxi Province China
- Department of Obstetrics and Gynecology Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang China
| | - Jinxue Zhang
- Orthopedic Department of Tangdu Hospital Fourth Military Medical University Xi'an Shaanxi Province China
| | - Yongming Liu
- Orthopedic Department of Tangdu Hospital Fourth Military Medical University Xi'an Shaanxi Province China
| | - Jingchang Ma
- Department of Immunology Fourth Military Medical University Xi'an Shaanxi Province China
| | - Dongxu Jiang
- Department of Immunology Fourth Military Medical University Xi'an Shaanxi Province China
| | - Xuexin Zhang
- Department of Immunology Fourth Military Medical University Xi'an Shaanxi Province China
- Department of Obstetrics and Gynecology Second Affiliated Hospital of Harbin Medical University Harbin Heilongjiang China
| | - Xin Yi
- Orthopedic Department of Tangdu Hospital Fourth Military Medical University Xi'an Shaanxi Province China
| | - Kun Cheng
- Transplant Immunology Laboratory Fourth Military Medical University Xi'an Shaanxi China
| | - Shen Shen
- Department of Immunology Fourth Military Medical University Xi'an Shaanxi Province China
- Transplant Immunology Laboratory Fourth Military Medical University Xi'an Shaanxi China
| | - Yixin Yang
- Department of Immunology Fourth Military Medical University Xi'an Shaanxi Province China
| | - Ran Zhuang
- Department of Immunology Fourth Military Medical University Xi'an Shaanxi Province China
- Transplant Immunology Laboratory Fourth Military Medical University Xi'an Shaanxi China
| | - Yuan Zhang
- Institute of Medical Research Northwest Polytechnic University Xi'an Shaanxi China
| |
Collapse
|
23
|
Kishi S, Brooks CR, Taguchi K, Ichimura T, Mori Y, Akinfolarin A, Gupta N, Galichon P, Elias BC, Suzuki T, Wang Q, Gewin L, Morizane R, Bonventre JV. Proximal tubule ATR regulates DNA repair to prevent maladaptive renal injury responses. J Clin Invest 2019; 129:4797-4816. [PMID: 31589169 PMCID: PMC6819104 DOI: 10.1172/jci122313] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/23/2019] [Indexed: 12/11/2022] Open
Abstract
Maladaptive proximal tubule (PT) repair has been implicated in kidney fibrosis through induction of cell-cycle arrest at G2/M. We explored the relative importance of the PT DNA damage response (DDR) in kidney fibrosis by genetically inactivating ataxia telangiectasia and Rad3-related (ATR), which is a sensor and upstream initiator of the DDR. In human chronic kidney disease, ATR expression inversely correlates with DNA damage. ATR was upregulated in approximately 70% of Lotus tetragonolobus lectin-positive (LTL+) PT cells in cisplatin-exposed human kidney organoids. Inhibition of ATR resulted in greater PT cell injury in organoids and cultured PT cells. PT-specific Atr-knockout (ATRRPTC-/-) mice exhibited greater kidney function impairment, DNA damage, and fibrosis than did WT mice in response to kidney injury induced by either cisplatin, bilateral ischemia-reperfusion, or unilateral ureteral obstruction. ATRRPTC-/- mice had more cells in the G2/M phase after injury than did WT mice after similar treatments. In conclusion, PT ATR activation is a key component of the DDR, which confers a protective effect mitigating the maladaptive repair and consequent fibrosis that follow kidney injury.
Collapse
Affiliation(s)
- Seiji Kishi
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Department of Nephrology, Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Japan
- Department of General Medicine, Kawasaki Medical School, Kurashiki, Japan
| | - Craig R. Brooks
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Kensei Taguchi
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Takaharu Ichimura
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Yutaro Mori
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Akinwande Akinfolarin
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Navin Gupta
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Pierre Galichon
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Sorbonne Université, INSERM UMR S1155, AP-HP, Hôpital Tenon, Paris, France
| | - Bertha C. Elias
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Tomohisa Suzuki
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Qian Wang
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Leslie Gewin
- Division of Nephrology and Hypertension, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Ryuji Morizane
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Joseph V. Bonventre
- Renal Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| |
Collapse
|
24
|
Abstract
As one type of the most common endogenous short noncoding RNAs (ncRNAs), microRNAs (miRNAs) act as posttranscriptional regulators of gene expression and have great potential biological functions in the physiological and pathological processes of various diseases. The role of miRNAs in renal fibrosis has also attracted great attention in the previous 20 years, and new therapeutic strategies targeting miRNAs appear to be promising. Some researchers have previously reviewed the roles of miRNA in renal fibrosis disease, but numerous studies have emerged over the recent 5 years. It is necessary to update and summarize research progress in miRNAs in renal fibrosis. Thus, in this review, we summarize progress in miRNA-mediated renal fibrosis over the last 5 years and evaluate the biological functions of some miRNAs in different stages of renal fibrosis. Furthermore, we also expound the recent clinical applications of these miRNAs to provide new insights into the treatment of renal fibrosis disease.
Collapse
Affiliation(s)
- Youling Fan
- Department of Anesthesiology, The First People's Hospital of Kashgar, Xinjiang Province, China.,Department of Anesthesiology, Panyu Central Hospital, Guangzhou, Guangdong Province, China
| | - Hongtao Chen
- Department of Anesthesiology, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong Province, China
| | - Zhenxing Huang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong Province, China
| | - Hong Zheng
- Department of Anesthesiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang Province, China
| | - Jun Zhou
- Department of Anesthesiology, The third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
25
|
Kajimura T, Sato S, Murakami A, Hayashi-Okada M, Nakashima K, Sueoka K, Sugino N. Overexpression of carbonyl reductase 1 inhibits malignant behaviors and epithelial mesenchymal transition by suppressing TGF-β signaling in uterine leiomyosarcoma cells. Oncol Lett 2019; 18:1503-1512. [PMID: 31423217 PMCID: PMC6607169 DOI: 10.3892/ol.2019.10429] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 05/22/2019] [Indexed: 12/31/2022] Open
Abstract
Carbonyl reductase 1 (CBR1) has been reported to be involved in cancer progression. Recently, we found that CBR1 overexpression inhibited malignant behaviors and the epithelial mesenchymal transition (EMT) in uterine cervical cancer. It remained unclear whether this was also the case in uterine leiomyosarcoma (uLMS), which is derived from mesenchymal cells and is a much more malignant gynecological tumor. A number of previous studies suggested that malignant behaviors are associated with EMT, even in mesenchymal malignant tumors. In the present study, we investigated whether CBR1 inhibits malignant behaviors and EMT in uLMS. We established clones of uLMS cells (SKN cells) and uterine sarcoma cells (MES-SA cells) that overexpressed CBR1. Cell proliferative, migratory and invasive activities were suppressed by CBR1 overexpression, accompanied by increases in the expressions of epithelial markers (E-cadherin and cytokeratin) and decreases in the expressions of mesenchymal markers (N-cadherin and fibronectin), suggesting that CBR1 overexpression inhibits malignant behaviors and EMT in uLMS cells. In addition, transforming growth factor-β (TGF-β) production and the subsequent signaling and phosphorylation of Smad were suppressed in the clones. To investigate the association between TGF-β and EMT, SKN cells were treated with TGF-β or a TGF-β receptor blocker (SB431542). EMT was promoted by TGF-β and inhibited by SB431542. In conclusion, this is the first study, to the best of the authors' knowledge, showing that CBR1 overexpression inhibits malignant behaviors and EMT in uLMS cells. The present study provided novel insight demonstrating that the suppressive effect of CBR1 is mediated through TGF-β signaling.
Collapse
Affiliation(s)
- Takuya Kajimura
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Shun Sato
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Akihiro Murakami
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Maki Hayashi-Okada
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Kengo Nakashima
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Kotaro Sueoka
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| | - Norihiro Sugino
- Department of Obstetrics and Gynecology, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi 755-8505, Japan
| |
Collapse
|
26
|
Sun IO, Lerman LO. Urinary microRNA in kidney disease: utility and roles. Am J Physiol Renal Physiol 2019; 316:F785-F793. [PMID: 30759023 PMCID: PMC6580242 DOI: 10.1152/ajprenal.00368.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/29/2019] [Accepted: 02/11/2019] [Indexed: 12/18/2022] Open
Abstract
MicroRNAs (miRNAs) are small, noncoding single-stranded RNA oligonucleotides that modulate physiological and pathological processes by modulating target gene expression. Many miRNAs display tissue-specific expression patterns, the dysregulation of which has been associated with various disease states, including kidney disease. Mounting evidence implicates miRNAs in various biological processes, such as cell proliferation and differentiation and cancer. Because miRNAs are relatively stable in tissue and biological fluids, particularly when carried by extracellular vesicles, changes in their levels may reflect the development of human disease. Urinary miRNAs originate from primary kidney and urinary tract cells, cells infiltrating the renal tissue and shed in the urine, or the systemic circulation. Although their validity as biomarkers for kidney disease has not been fully established, studies have been applying analysis of miRNAs in the urine in an attempt to detect and monitor acute and chronic renal diseases. Because appreciation of the significance of miRNAs in the renal field is on the rise, an understanding of miRNA pathways that regulate renal physiology and pathophysiology is becoming critically important. This review aims to summarize new data obtained in this field of research. It is hoped that new developments in the use of miRNAs as biomarkers and/or therapy will help manage and contain kidney disease in affected subjects.
Collapse
Affiliation(s)
- In O Sun
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
- Division of Nephrology, Department of Internal Medicine, Presbyterian Medical Center, Jeonju, Korea
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| |
Collapse
|
27
|
Non-Coding RNAs as New Therapeutic Targets in the Context of Renal Fibrosis. Int J Mol Sci 2019; 20:ijms20081977. [PMID: 31018516 PMCID: PMC6515288 DOI: 10.3390/ijms20081977] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/19/2019] [Accepted: 04/20/2019] [Indexed: 12/15/2022] Open
Abstract
Fibrosis, or tissue scarring, is defined as the excessive, persistent and destructive accumulation of extracellular matrix components in response to chronic tissue injury. Renal fibrosis represents the final stage of most chronic kidney diseases and contributes to the progressive and irreversible decline in kidney function. Limited therapeutic options are available and the molecular mechanisms governing the renal fibrosis process are complex and remain poorly understood. Recently, the role of non-coding RNAs, and in particular microRNAs (miRNAs), has been described in kidney fibrosis. Seminal studies have highlighted their potential importance as new therapeutic targets and innovative diagnostic and/or prognostic biomarkers. This review will summarize recent scientific advances and will discuss potential clinical applications as well as future research directions.
Collapse
|
28
|
Zhou H, Gao L, Yu Z, Hong S, Zhang Z, Qiu Z. LncRNA HOTAIR promotes renal interstitial fibrosis by regulating Notch1 pathway via the modulation of miR‐124. Nephrology (Carlton) 2019; 24:472-480. [PMID: 29717517 DOI: 10.1111/nep.13394] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/23/2018] [Indexed: 01/17/2023]
Affiliation(s)
- Hao Zhou
- Department of UrologyThe Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine (The People's Hospital of Fujian Province) Fuzhou China
| | - Lin Gao
- Department of UrologyThe Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine (The People's Hospital of Fujian Province) Fuzhou China
| | - Zuo‐hua Yu
- Department of UrologyThe Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine (The People's Hospital of Fujian Province) Fuzhou China
| | - Shi‐jun Hong
- Department of UrologyThe Affiliated People's Hospital of Fujian University of Traditional Chinese Medicine (The People's Hospital of Fujian Province) Fuzhou China
| | - Zhi‐wei Zhang
- Department of ResearchBeijing Zhong Jian Dong Ke Company Beijing China
| | - Zhen‐zhen Qiu
- Department of Physical EducationMinjiang University Fuzhou China
| |
Collapse
|
29
|
Gholaminejad A, Abdul Tehrani H, Gholami Fesharaki M. Identification of candidate microRNA biomarkers in renal fibrosis: a meta-analysis of profiling studies. Biomarkers 2018; 23:713-724. [PMID: 29909697 DOI: 10.1080/1354750x.2018.1488275] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Alieh Gholaminejad
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hossein Abdul Tehrani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
30
|
Liao YW, Yu CC, Hsieh PL, Chang YC. miR-200b ameliorates myofibroblast transdifferentiation in precancerous oral submucous fibrosis through targeting ZEB2. J Cell Mol Med 2018; 22:4130-4138. [PMID: 29893466 PMCID: PMC6111815 DOI: 10.1111/jcmm.13690] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 04/20/2018] [Indexed: 12/28/2022] Open
Abstract
Oral submucous fibrosis (OSF) is a progressive scarring disease. MicroRNA-200b (miR-200b) has been reported as a tumour suppressor, but its role in the precancerous OSF remains unknown. In this study, we investigated the impact of miR-200b on myofibroblastic differentiation activity. Arecoline is a major areca nut alkaloid and has been employed to induce the elevated myofibroblast activity in human buccal mucosal fibroblasts (BMFs). Treatment of arecoline in BMFs dose-dependently reduced gene expression of miR-200b, which corresponded with the decreased expression of miR-200b in fBMFs. The arecoline-induced myofibroblast activities were abolished by overexpression of miR-200b in BMFs, and the same results were observed in fBMFs. In addition, α-SMA was inhibited by an increase in miR-200b. We further demonstrated that miR-200b-mediated decrease in ZEB2 led to down-regulation of α-SMA, vimentin. Loss of miR-200b resulted in enhanced collagen contraction and migration capabilities, and knockdown of ZEB2 reversed these phenomena. Lastly, we showed the expression of miR-200b was significantly less and ZEB2 was markedly higher in OSF tissues. These results suggested that down-regulation of miR-200b may contribute to the pathogenesis of areca quid-associated OSF through the regulation of ZEB2 and myofibroblast hallmarks.
Collapse
Affiliation(s)
- Yi-Wen Liao
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan
| | - Cheng-Chia Yu
- Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan.,Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ling Hsieh
- Institute of Oral Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chao Chang
- School of Dentistry, Chung Shan Medical University, Taichung, Taiwan.,Department of Dentistry, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
31
|
Chen L, Chen R, Kemper S, Cong M, You H, Brigstock DR. Therapeutic effects of serum extracellular vesicles in liver fibrosis. J Extracell Vesicles 2018; 7:1461505. [PMID: 29696080 PMCID: PMC5912192 DOI: 10.1080/20013078.2018.1461505] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 04/02/2018] [Indexed: 12/15/2022] Open
Abstract
The lack of approved therapies for hepatic fibrosis seriously limits medical management of patients with chronic liver disease. Since extracellular vesicles (EVs) function as conduits for intercellular molecular transfer, we investigated if EVs from healthy individuals have anti-fibrotic properties. Hepatic fibrogenesis or fibrosis in carbon tetrachloride (CCl4)- or thioacetic acid-induced liver injury models in male or female mice were suppressed by serum EVs from normal mice (EVN) but not from fibrotic mice (EVF). CCl4-treated mice undergoing EVN therapy also exhibited reduced levels of hepatocyte death, inflammatory infiltration, circulating AST/ALT levels and hepatic or circulating pro-inflammatory cytokines. Hepatic histology, liver function tests or circulating proinflammatory cytokine levels were unaltered in control mice receiving EVN. As determined using PKH26-labelled EVN, principal target cells included hepatic stellate cells (HSC; a normally quiescent fibroblastic cell that undergoes injury-induced activation and produces fibrosis during chronic injury) or hepatocytes which showed increased EVN binding after, respectively, activation or exposure to CCl4. In vitro, EVN decreased proliferation and fibrosis-associated molecule expression in activated HSC, while reversing the inhibitory effects of CCl4 or ethanol on hepatocyte proliferation. In mice, microRNA-34c, -151-3p, -483-5p, -532-5p and -687 were more highly expressed in EVN than EVF and mimics of these microRNAs (miRs) individually suppressed fibrogenic gene expression in activated HSC. A role for these miRs in contributing to EVN actions was shown by the ability of their corresponding antagomirs to individually and/or collectively block the therapeutic effects of EVN on activated HSC or injured hepatocytes. Similarly, the activated phenotype of human LX-2 HSC was attenuated by serum EVs from healthy human subjects and contained higher miR-34c, -151-3p, -483-5p or -532-5p than EVs from hepatic fibrosis patients. In conclusion, serum EVs from normal healthy individuals are inherently anti-fibrogenic and anti-fibrotic, and contain microRNAs that have therapeutic actions in activated HSC or injured hepatocytes. Abbreviations: ALT: alanine aminotransferase; AST: aspartate aminotransferase; CCl4: carbon tetrachloride; CCN2: connective tissue growth factor; E: eosin; EGFP: enhanced green fluorescent protein; EVs: extracellular vesicles; EVF: serum EVs from mice with experimental hepatic fibrosis; EVN: serum EVs from normal mice; H: hematoxylin; HSC: hepatic stellate cell; IHC: immunohistochemistry; IL: interleukin; MCP-1: monocyte chemotactic protein-1; miR: microRNA; mRNA: messenger RNA; NTA: nanoparticle tracking analysis; PCNA: proliferating cell nuclear antigen; qRT-PCR: quantitative real-time polymerase chain reaction; SDS-PAGE: sodium dodecyl sulphate – polyacrylamide gel electrophoresis; αSMA: alpha smooth muscle actin; TAA: thioacetic acid; TG: transgenic; TGF-β: transforming growth factor beta; TEM: transmission electron microscopy; TNFα: tumour necrosis factor alpha.
Collapse
Affiliation(s)
- Li Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Ruju Chen
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Sherri Kemper
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Min Cong
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - Hong You
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, National Clinical Research Center of Digestive Diseases, Beijing, China
| | - David R Brigstock
- Center for Clinical and Translational Research, The Research Institute at Nationwide Children's Hospital, Columbus, OH, USA.,Department of Surgery, Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
32
|
Meder L, König K, Dietlein F, Macheleidt I, Florin A, Ercanoglu MS, Rommerscheidt-Fuss U, Koker M, Schön G, Odenthal M, Klein F, Büttner R, Schulte JH, Heukamp LC, Ullrich RT. LIN28B enhanced tumorigenesis in an autochthonous KRAS G12V-driven lung carcinoma mouse model. Oncogene 2018; 37:2746-2756. [PMID: 29503447 DOI: 10.1038/s41388-018-0158-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 11/21/2017] [Accepted: 01/10/2018] [Indexed: 12/30/2022]
Abstract
LIN28B is a RNA-binding protein regulating predominantly let-7 microRNAs with essential functions in inflammation, wound healing, embryonic stem cells, and cancer. LIN28B expression is associated with tumor initiation, progression, resistance, and poor outcome in several solid cancers, including lung cancer. However, the functional role of LIN28B, especially in non-small cell lung adenocarcinomas, remains elusive. Here, we investigated the effects of LIN28B expression on lung tumorigenesis using LIN28B transgenic overexpression in an autochthonous KRASG12V-driven mouse model. We found that LIN28B overexpression significantly increased the number of CD44+/CD326+ tumor cells, upregulated VEGF-A and miR-21 and promoted tumor angiogenesis and epithelial-to-mesenchymal transition (EMT) accompanied by enhanced AKT phosphorylation and nuclear translocation of c-MYC. Moreover, LIN28B accelerated tumor initiation and enhanced proliferation which led to a shortened overall survival. In addition, we analyzed lung adenocarcinomas of the Cancer Genome Atlas (TCGA) and found LIN28B expression in 24% of KRAS-mutated cases, which underscore the relevance of our model.
Collapse
Affiliation(s)
- Lydia Meder
- Department I of Internal Medicine, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany. .,Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany.
| | - Katharina König
- Labor Dr. Quade und Kollegen GmbH, Aachener Straße 338, Cologne, 50933, Germany
| | - Felix Dietlein
- Department of Medical Oncology, Dana-Faber Cancer Institute, Boston, MA, 02215, USA.,Cancer Program, Broad Institute of MIT and Havard, Cambridge, MA, 02142, USA
| | - Iris Macheleidt
- Institute for Pathology, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Alexandra Florin
- Institute for Pathology, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Meryem S Ercanoglu
- Institute of Virology, Laboratory of Experimental Immunology, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany
| | | | - Mirjam Koker
- Department I of Internal Medicine, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany
| | - Gisela Schön
- Department I of Internal Medicine, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany
| | - Margarete Odenthal
- Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany.,Institute for Pathology, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Florian Klein
- Institute of Virology, Laboratory of Experimental Immunology, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany
| | - Reinhard Büttner
- Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany.,Institute for Pathology, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany.,Center for Integrated Oncology Cologne/Bonn, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany
| | - Johannes H Schulte
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Augustenburger Platz 1, Berlin, 13353, Germany.,German Cancer Consortium (DKTK Berlin), Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany.,Deutsches Krebsforschungszentrum Heidelberg (DKFZ), Heidelberg, Germany
| | - Lukas C Heukamp
- New Oncology, Gottfried-Hagen-Straße 20, 51105, Cologne, Germany.,Institute for Hematopathology Hamburg, Fangdieckstraße 75a, Hamburg, Germany
| | - Roland T Ullrich
- Department I of Internal Medicine, University Hospital Cologne, Kerpener Straße 62, Cologne, 50937, Germany.,Center for Molecular Medicine Cologne, University of Cologne, Robert-Koch Straße 21, Cologne, 50931, Germany
| |
Collapse
|
33
|
Nishimoto Y, Murakami A, Sato S, Kajimura T, Nakashima K, Yakabe K, Sueoka K, Sugino N. Decreased carbonyl reductase 1 expression promotes tumor growth via epithelial mesenchymal transition in uterine cervical squamous cell carcinomas. Reprod Med Biol 2018; 17:173-181. [PMID: 29692675 PMCID: PMC5902461 DOI: 10.1002/rmb2.12086] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Accepted: 12/25/2017] [Indexed: 11/09/2022] Open
Abstract
Purpose Carbonyl reductase 1 (CBR1) is involved in cancer progression. Recently, the authors reported that the loss of CBR1 expression is associated with a poor prognosis in uterine cervical cancer. Here, we investigated whether the decreased CBR1 expression promotes cancer progression by inducing the epithelial mesenchymal transition (EMT). Methods Antisense constructs of CBR1 complementary DNA (antisense clones) and the empty vectors (control clones) were transfected into human uterine cervical squamous cell carcinoma cell lines (SKG II and SiHa) and the proliferation and EMT marker expression of these clones were analyzed in vitro. In an in vivo study, 107 cells of the antisense and control clones were subcutaneously injected into nude mice and the tumorigenesis was observed for 8 weeks. Results With the decreased CBR1 expression, the proliferation of the antisense clones increased, accompanied by a decrease in epithelial markers (E-cadherin and cytokeratin) and an increase in mesenchymal markers (fibronectin, alpha-smooth muscle actin, and N-cadherin), which suggests EMT induction. In the in vivo study, the tumor volume in the antisense group was significantly larger than that in the control group. Conclusion Decreased CBR1 expression promotes tumor growth by inducing EMT in uterine cervical squamous cell carcinomas.
Collapse
Affiliation(s)
- Yuki Nishimoto
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| | - Akihiro Murakami
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| | - Shun Sato
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| | - Takuya Kajimura
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| | - Kengo Nakashima
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| | - Kazuyuki Yakabe
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| | - Kotaro Sueoka
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| | - Norihiro Sugino
- Department of Obstetrics and Gynecology Yamaguchi University Graduate School of Medicine Ube Japan
| |
Collapse
|
34
|
Ichii O, Horino T. MicroRNAs associated with the development of kidney diseases in humans and animals. J Toxicol Pathol 2018; 31:23-34. [PMID: 29479137 PMCID: PMC5820100 DOI: 10.1293/tox.2017-0051] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 09/10/2017] [Indexed: 12/20/2022] Open
Abstract
Mature microRNAs (miRNAs) are single-stranded RNAs with approximately 18-25 bases, and their sequences are highly conserved among animals. miRNAs act as posttranscriptional regulators by binding mRNAs, and their main function involves the degradation of their target mRNAs. Recent studies revealed altered expression of miRNAs in the kidneys during the progression of acute kidney injury (AKI) and chronic kidney disease (CKD) in humans and experimental rodent models by using high-throughput screening techniques including microarray and small RNA sequencing. Particularly, miR-21 seems to be strongly associated with renal pathogenesis both in the glomerulus and tubulointerstitium. Furthermore, abundant evidence has been gathered showing the involvement of miRNAs in renal fibrosis. Because of the complex morphofunctional organization of the mammalian kidneys, it is crucial both to determine the exact localization of the kidney cells that express the miRNAs, which has been addressed mainly using in situ hybridization methods, and to identify precisely which mRNAs are bound and degraded by these miRNAs, which has been studied mostly through in vitro analysis. To discover novel biomarker candidates, miRNA levels in urine supernatant, sediment, and exosomal fraction were comprehensively investigated in different types of kidney disease, including drug-induced AKI, ischemia-induced AKI, diabetic nephropathy, lupus nephritis, and IgA nephropathy. Recent studies also demonstrated the therapeutic effect of miRNA and/or anti-miRNA administrations. The intent of this review is to illustrate the state-of-the-art research in the field of miRNAs associated with renal pathogenesis, especially focusing on AKI and CKD in humans and animal models.
Collapse
Affiliation(s)
- Osamu Ichii
- Laboratory of Anatomy, Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18-Nishi 9, Kita-ku,
Sapporo, Hokkaido 060-0818, Japan
| | - Taro Horino
- Laboratory of Anatomy, Department of Basic Veterinary
Sciences, Faculty of Veterinary Medicine, Hokkaido University, Kita 18-Nishi 9, Kita-ku,
Sapporo, Hokkaido 060-0818, Japan
| |
Collapse
|
35
|
Wang W, Zhou PH, Hu W, Xu CG, Zhou XJ, Liang CZ, Zhang J. Cryptotanshinone hinders renal fibrosis and epithelial transdifferentiation in obstructive nephropathy by inhibiting TGF-β1/Smad3/integrin β1 signal. Oncotarget 2017; 9:26625-26637. [PMID: 29928474 PMCID: PMC6003568 DOI: 10.18632/oncotarget.23803] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 09/03/2017] [Indexed: 01/28/2023] Open
Abstract
Recent studies have reported that CTS can alleviate cardiac fibrosis. However, the effects of CTS on kidney fibrosis and EMT are still unknown. This study explored whether CTS could attenuate tubulointerstitial fibrosis as well as EMT, and investigated the potential underlying mechanisms. In this study, an in vivo UUO mouse model and an in vitro TGF-β1 stimulated normal renal tubular kidney epithelial cell model were established. In UUO model, administration of 50 mg kg-1 day-1 CTS markedly decreased the occurrence of kidney injury and the accumulation of fibronectin and collagen-1. In addition, CTS reduced the expression level of α-SMA but retained E-cadherin in obstructed kidneys. In vitro, CTS suppressed the expression of fibronectin, collagen-1 and α-SMA but retained that of E-cadherin. Furthermore, CTS selectively abolished the activation of Smad3 and suppressed the nuclear translocation of Smad2, Smad3 and Smad4. CTS could block the promoter activity of integrin β1 induced by Smad3. Furthermore, CTS inhibited Smad3 binding to integrin β1 promoter sequences. These data suggest that CTS can ameliorate kidney fibrosis and EMT, at least in part, by inhibiting the TGF-β1/Smad3/integrin β1 signaling pathway.
Collapse
Affiliation(s)
- Wei Wang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, 232200, China.,Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Pang-Hu Zhou
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Wei Hu
- Department of Urology, The First Affiliated Hospital of Nan-Hua University, Henyang, Hunan, 421001, China
| | - Chang-Geng Xu
- Department of Urology, Wuhan Central Hospital, Wuhan, Hubei Province, 430014, China
| | - Xiang-Jun Zhou
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Chao-Zhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology, Anhui Medical University, Hefei, Anhui, 232200, China
| | - Jie Zhang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China.,Huangshi Central Hospital, Hubei Polytechnic University, Huangshi, Hubei Province, 435000, China
| |
Collapse
|
36
|
Genome-wide Profiling of Urinary Extracellular Vesicle microRNAs Associated With Diabetic Nephropathy in Type 1 Diabetes. Kidney Int Rep 2017; 3:555-572. [PMID: 29854963 PMCID: PMC5976846 DOI: 10.1016/j.ekir.2017.11.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 11/15/2017] [Accepted: 11/27/2017] [Indexed: 01/01/2023] Open
Abstract
Introduction Diabetic nephropathy (DN) is a form of progressive kidney disease that often leads to end-stage renal disease (ESRD). It is initiated by microvascular complications due to diabetes. Although microalbuminuria (MA) is the earliest clinical indication of DN among patients with type 1 diabetes (T1D), it lacks the sensitivity and specificity to detect the early onset of DN. Recently, microRNAs (miRNAs) have emerged as critical regulators in diabetes as well as various forms of kidney disease, including renal fibrosis, acute kidney injury, and progressive kidney disease. Additionally, circulating extracellular miRNAs, especially miRNAs packaged in extracellular vesicles (EVs), have garnered significant attention as potential noninvasive biomarkers for various diseases and health conditions. Methods As part of the University of Pittsburgh Epidemiology of Diabetes Complications (EDC) study, urine was collected from individuals with T1D with various grades of DN or MA (normal, overt, intermittent, and persistent) over a decade at prespecified intervals. We isolated EVs from urine and analyzed the small-RNA using NextGen sequencing. Results We identified a set of miRNAs that are enriched in urinary EVs compared with EV-depleted samples, and identified a number of miRNAs showing concentration changes associated with DN occurrence, MA status, and other variables, such as hemoglobin A1c levels. Conclusion Many of the miRNAs associated with DN occurrence or MA status directly target pathways associated with renal fibrosis (including transforming growth factor-β and phosphatase and tensin homolog), which is one of the major contributors to the pathology of DN. These miRNAs are potential biomarkers for DN and MA.
Collapse
|
37
|
Lv W, Fan F, Wang Y, Gonzalez-Fernandez E, Wang C, Yang L, Booz GW, Roman RJ. Therapeutic potential of microRNAs for the treatment of renal fibrosis and CKD. Physiol Genomics 2017; 50:20-34. [PMID: 29127220 DOI: 10.1152/physiolgenomics.00039.2017] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Chronic kidney disease (CKD), defined as reduced glomerular filtration rate, is increasingly becoming a major public health issue. At the histological level, renal fibrosis is the final common pathway leading to end-stage renal disease, irrespective of the initial injury. According to this view, antifibrotic agents should slow or halt the progression of CKD. However, due to multiple overlapping pathways stimulating fibrosis, it has been difficult to develop antifibrotic drugs that delay or reverse the progression of CKD. MicroRNAs (miRNAs) are small noncoding RNA molecules, 18-22 nucleotides in length, that control many developmental and cellular processes as posttranscriptional regulators of gene expression. Emerging evidence suggests that miRNAs targeted against genes involved in renal fibrosis might be potential candidates for the development of antifibrotic therapies for CKD. This review will discuss some of the miRNAs, such as Let-7, miR-21,-29, -192, -200,-324, -132, -212, -30, -126, -433, -214, and -199a, that are implicated in renal fibrosis and the potential to exploit these molecular targets for the treatment of CKD.
Collapse
Affiliation(s)
- Wenshan Lv
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, Mississippi.,Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University , Qingdao , China
| | - Fan Fan
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Yangang Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University , Qingdao , China
| | - Ezekiel Gonzalez-Fernandez
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, Mississippi
| | - Chen Wang
- Department of Endocrinology and Metabolism, the Affiliated Hospital of Qingdao University , Qingdao , China
| | - Lili Yang
- West Coast Clinic of Affiliated Hospital of Qingdao University , Qingdao , China
| | - George W Booz
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center , Jackson, Mississippi
| | | |
Collapse
|
38
|
Park EJ, Jung HJ, Choi HJ, Cho JI, Park HJ, Kwon TH. miR-34c-5p and CaMKII are involved in aldosterone-induced fibrosis in kidney collecting duct cells. Am J Physiol Renal Physiol 2017; 314:F329-F342. [PMID: 29070573 DOI: 10.1152/ajprenal.00358.2017] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mineralocorticoids trigger a profibrotic process in the kidney. In mouse cortical collecting duct cells, the present study addressed two main questions: 1) what are microRNAs (miRNAs) and their target genes that are changed by aldosterone? and 2) what do miRNAs, in response to aldosterone, regulate regarding signaling pathways related to fibrosis? A microarray chip assay was done in cells in the absence or presence of aldosterone treatment (10-6 M; 3 days). The candidate miRNAs were identified by the criteria of >30% of fold change among the significantly changed miRNAs ( P < 0.05). Twenty-nine miRNAs were upregulated (>1.3-fold), and 27 miRNAs were downregulated (<0.7-fold). Putative target genes of identified miRNAs were associated with 74 Kyoto Encyclopedia of Genes and Genomes pathways. Among them, the wingless-related integration site (Wnt) signaling pathway was highly ranked, where 15 mature miRNAs were observed. These miRNAs were further analyzed by real-time quantitative PCR, and among them, miR-130b-3p, miR-34c-5p, and miR-146a-5p were selected. Through the identification of putative target genes of these three miRNAs, mRNA and protein expression of the Ca2+/calmodulin-dependent protein kinase type II β-chain ( Camk2b) gene (a target gene of miR-34c-5p) were found to be increased significantly in aldosterone-treated cells, where fibronectin (FN) and α-smooth muscle actin were induced. When CaMKIIβ small interfering RNA or the miR-34c-5p mimic was transfected, aldosterone-induced FN expression was significantly attenuated, along with reduced CaMKIIβ protein expression. A luciferase reporter assay revealed a decrease of CaMKIIβ translation in cells transfected with miRNA mimics of miR-34c-5p. In conclusion, aldosterone-induced downregulation of miR-34c-5p in the Wnt signaling pathway and a consequent increase of CaMKIIβ expression are likely to be involved in aldosterone-induced fibrosis.
Collapse
Affiliation(s)
- Eui-Jung Park
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Taegu, Korea
| | - Hyun Jun Jung
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea.,Epithelial Systems Biology Laboratory, Systems Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health , Bethesda, Maryland
| | - Hyo-Jung Choi
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea
| | - Jeong-In Cho
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Taegu, Korea
| | - Hye-Jeong Park
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Taegu, Korea
| | - Tae-Hwan Kwon
- Department of Biochemistry and Cell Biology, School of Medicine, Kyungpook National University, Taegu, Korea.,BK21 Plus KNU Biomedical Convergence Program, Department of Biomedical Science, School of Medicine, Kyungpook National University, Taegu, Korea
| |
Collapse
|
39
|
Xin G, Zhou G, Zhang X, Wang W. Potential role of upregulated microRNA‑146b and ‑21 in renal fibrosis. Mol Med Rep 2017; 16:2863-2867. [PMID: 28714020 DOI: 10.3892/mmr.2017.6929] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 05/09/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to identify candidate microRNAs (miRNAs) involved in the progression of renal fibrosis. Dataset GSE42716 of miRNAs extracted from kidneys from mice with unilateral ureteral obstruction (UUO), and mice without UUO was downloaded from the Gene Expression Omnibus database. The Limma package was used to identify differential expression between mice with and without UUO. Renal disease‑related miRNAs were predicted based on the miRWalk database. Thereafter, candidate miRNAs were screened by taking the intersection of differentially expressed miRNAs and predicted miRNAs, followed by screening of target genes using miRWalk and transcription factors using the TransmiR database. An integrative regulatory network was constructed using Cytoscape software. Enrichment analysis of target genes was also performed. In total, 76 differentially expressed miRNAs were identified in kidneys with UUO compared with the normal samples based on dataset and 9 miRNAs were identified as related to renal disease from the miRWalk database. A Venn diagram revealed two overlapping upregulated miRNAs; miR‑146b and miR‑21. Transcription factor NFKB1 may activate miR‑146b and AKT may activate miR‑21. In addition, miR‑21 had a regulatory effect on IFNG expression and miR‑146b may regulate the expression of BCL2, PTEN and IFNG. Furthermore, target genes of miR‑146b and miR‑21 were significantly enriched in 14 Gene Ontology terms including regulation of cell proliferation (e.g., BCL2, PTEN and IFNG). Overexpression of miR‑21 may be activated by AKT2 and contribute to renal fibrosis by negatively regulating IFNG expression. Furthermore, miR‑146b may be activated by NFKB1 and subsequently reduce the expression of BCL2, PTEN and IFNG in the progression of renal fibrosis.
Collapse
Affiliation(s)
- Guangda Xin
- Department of Nephrology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Guangyu Zhou
- Department of Nephrology, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xiaofei Zhang
- Department of Pediatrics, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Wanning Wang
- Department of Nephrology, The First Bethune Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
40
|
Wang D, Xin L, Lin JH, Liao Z, Ji JT, Du TT, Jiang F, Li ZS, Hu LH. Identifying miRNA-mRNA regulation network of chronic pancreatitis based on the significant functional expression. Medicine (Baltimore) 2017; 96:e6668. [PMID: 28538367 PMCID: PMC5457847 DOI: 10.1097/md.0000000000006668] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND The aim of this study was to explore the underlying molecular mechanism and potential molecular biomarkers of chronic pancreatitis (CP) and construct a miRNA-mRNA regulation network. METHODS To explore the involvement of miRNAs in CP, we downloaded the miRNA and mRNA expression profiles of CP patients and healthy controls and identified the differentially expressed miRNAs and genes. Functional analysis was conducted and significant pathways were utilized. Finally, the miRNA-mRNA regulation network of CP was constructed. RESULTS A total of 44 miRNA risk gene pathway relationships were identified, and a complex regulation network was constructed with 3 genes (ABL1, MYC, and ANAPC13) having the highest degree in affecting the network of CP. Importantly, 4 risk genes (NOTCH3, COX5A, THBS1, and KARS) and 1 risk miRNA (hsa-miR-324-5p) were identified with high prediction accuracy. CONCLUSIONS In conclusion, we analyzed miRNAs and mRNAs expression profiles in CP, 1 risk miRNA, and 4 risk genes were identified with high prediction accuracy as biomarkers of CP. Although further evaluation in clinical study is needed, our findings provide new insights into the pathogenesis of CP and may improve the diagnosis and therapy by identifying novel targets.
Collapse
Affiliation(s)
| | - Lei Xin
- Digestive Endoscopy Center, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | | | - Zhuan Liao
- Digestive Endoscopy Center, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | | | | | - Fei Jiang
- Digestive Endoscopy Center, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Zhao-Shen Li
- Digestive Endoscopy Center, Changhai Hospital, the Second Military Medical University, Shanghai, China
| | | |
Collapse
|
41
|
Wen L, Andersen PK, Husum DMU, Nørregaard R, Zhao Z, Liu Z, Birn H. MicroRNA-148b regulates megalin expression and is associated with receptor downregulation in mice with unilateral ureteral obstruction. Am J Physiol Renal Physiol 2017; 313:F210-F217. [PMID: 28331063 DOI: 10.1152/ajprenal.00585.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 03/02/2017] [Accepted: 03/16/2017] [Indexed: 11/22/2022] Open
Abstract
Megalin is a multiligand, endocytic receptor that is important for the normal, proximal tubule reabsorption of filtered proteins, hormones, enzymes, essential nutrients, and nephrotoxins. Megalin dysfunction has been associated with acute, as well as chronic kidney diseases. Tubular proteinuria has been observed following unilateral ureteral obstruction (UUO), suggesting megalin dysfunction; however, the pathophysiological mechanism has not been determined. To identify potential regulators of megalin expression, we examined renal microRNAs (miRNAs) expression and observed an upregulation of microRNA-148b (miR-148b) in obstructed mouse kidneys 7 days after UUO, which was associated with a significant reduction in proximal tubule megalin expression and accumulation of megalin ligands. By in silico miRNA target prediction analysis, we identified megalin messenger RNA (mRNA) as a potential target of miR-148b and confirmed using a dual-luciferase reporter assay that miR-148b targeted the 3'-untranslated region of the megalin gene. Transfection of LLC-PK1 cells with miR-148b mimic reduced endogenous megalin mRNA and protein levels in a concentration-dependent manner, while transfection with miR-148b inhibitor resulted in an increase. Our findings suggest that miR-148b directly downregulates megalin expression and that miR-148b negatively regulates megalin expression in UUO-induced kidney injury. Furthermore, the identification of a miRNA regulating megalin expression may allow for targeted interventions to modulate megalin function and proximal tubule uptake of proteins, as well as other ligands.
Collapse
Affiliation(s)
- Lu Wen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark.,Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Pia K Andersen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Dina M U Husum
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; and
| | - Rikke Nørregaard
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark; and
| | - Zhanzheng Zhao
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhangsuo Liu
- Department of Nephrology, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Henrik Birn
- Department of Biomedicine, Aarhus University, Aarhus, Denmark; .,Department of Renal Medicine, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
42
|
Venø MT, Venø ST, Rehberg K, van Asperen JV, Clausen BH, Holm IE, Pasterkamp RJ, Finsen B, Kjems J. Cortical Morphogenesis during Embryonic Development Is Regulated by miR-34c and miR-204. Front Mol Neurosci 2017; 10:31. [PMID: 28232790 PMCID: PMC5299138 DOI: 10.3389/fnmol.2017.00031] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 01/26/2017] [Indexed: 01/26/2023] Open
Abstract
The porcine brain closely resembles the human brain in aspects such as development and morphology. Temporal miRNA profiling in the developing embryonic porcine cortex revealed a distinct set of miRNAs, including miR-34c and miR-204, which exhibited a highly specific expression profile across the time of cortical folding. These miRNAs were found to target Doublecortin (DCX), known to be involved in neuron migration during cortical folding of gyrencephalic brains. In vivo modulation of miRNA expression in mouse embryos confirmed that miR-34c and miR-204 can control neuronal migration and cortical morphogenesis, presumably by posttranscriptional regulation of DCX.
Collapse
Affiliation(s)
- Morten T Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center, Aarhus University Aarhus, Denmark
| | - Susanne T Venø
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center, Aarhus University Aarhus, Denmark
| | - Kati Rehberg
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Jessy V van Asperen
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Bettina H Clausen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Ida E Holm
- Laboratory for Experimental Neuropathology, Department of Pathology, Randers Hospital Randers, Denmark
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht Utrecht, Netherlands
| | - Bente Finsen
- Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark Odense, Denmark
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Interdisciplinary Nanoscience Center, Aarhus University Aarhus, Denmark
| |
Collapse
|
43
|
Abstract
The significant parallels between cell plasticity during embryonic development and carcinoma progression have helped us understand the importance of the epithelial-mesenchymal transition (EMT) in human disease. Our expanding knowledge of EMT has led to a clarification of the EMT program as a set of multiple and dynamic transitional states between the epithelial and mesenchymal phenotypes, as opposed to a process involving a single binary decision. EMT and its intermediate states have recently been identified as crucial drivers of organ fibrosis and tumor progression, although there is some need for caution when interpreting its contribution to metastatic colonization. Here, we discuss the current state-of-the-art and latest findings regarding the concept of cellular plasticity and heterogeneity in EMT. We raise some of the questions pending and identify the challenges faced in this fast-moving field.
Collapse
|
44
|
Yamaguchi S, Morizane R, Homma K, Monkawa T, Suzuki S, Fujii S, Koda M, Hiratsuka K, Yamashita M, Yoshida T, Wakino S, Hayashi K, Sasaki J, Hori S, Itoh H. Generation of kidney tubular organoids from human pluripotent stem cells. Sci Rep 2016; 6:38353. [PMID: 27982115 PMCID: PMC5159864 DOI: 10.1038/srep38353] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/08/2016] [Indexed: 12/12/2022] Open
Abstract
Recent advances in stem cell research have resulted in methods to generate kidney organoids from human pluripotent stem cells (hPSCs), which contain cells of multiple lineages including nephron epithelial cells. Methods to purify specific types of cells from differentiated hPSCs, however, have not been established well. For bioengineering, cell transplantation, and disease modeling, it would be useful to establish those methods to obtain pure populations of specific types of kidney cells. Here, we report a simple two-step differentiation protocol to generate kidney tubular organoids from hPSCs with direct purification of KSP (kidney specific protein)-positive cells using anti-KSP antibody. We first differentiated hPSCs into mesoderm cells using a glycogen synthase kinase-3β inhibitor for 3 days, then cultured cells in renal epithelial growth medium to induce KSP+ cells. We purified KSP+ cells using flow cytometry with anti-KSP antibody, which exhibited characteristics of all segments of kidney tubular cells and cultured KSP+ cells in 3D Matrigel, which formed tubular organoids in vitro. The formation of tubular organoids by KSP+ cells induced the acquisition of functional kidney tubules. KSP+ cells also allowed for the generation of chimeric kidney cultures in which human cells self-assembled into 3D tubular structures in combination with mouse embryonic kidney cells.
Collapse
Affiliation(s)
- Shintaro Yamaguchi
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ryuji Morizane
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Renal Division, Brigham and Women's Hospital, 75 Francis Street, Boston, MA 02115, USA.,Department of Medicine, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA.,Harvard Stem Cell Institute, 7 Divinity Ave, Cambridge, MA 02138, USA
| | - Koichiro Homma
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Toshiaki Monkawa
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan.,Medical Education Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Sayuri Suzuki
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shizuka Fujii
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Muneaki Koda
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ken Hiratsuka
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Maho Yamashita
- Apheresis and Dialysis Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Tadashi Yoshida
- Apheresis and Dialysis Center, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shu Wakino
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Koichi Hayashi
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Junichi Sasaki
- Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Shingo Hori
- Emergency and Critical Care Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Hiroshi Itoh
- Department of Internal Medicine, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
45
|
Zou XZ, Liu T, Gong ZC, Hu CP, Zhang Z. MicroRNAs-mediated epithelial-mesenchymal transition in fibrotic diseases. Eur J Pharmacol 2016; 796:190-206. [PMID: 27916556 DOI: 10.1016/j.ejphar.2016.12.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 11/24/2016] [Accepted: 12/01/2016] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs), a large family of small and highly conserved non-coding RNAs, regulate gene expression through translational repression or mRNA degradation. Aberrant expression of miRNAs underlies a spectrum of diseases including organ fibrosis. Recent evidence suggests that miRNAs contribute to organ fibrosis through mediating epithelial-mesenchymal transition (EMT). Alleviation of EMT has been proposed as a promising strategy against fibrotic diseases given the key role of EMT in fibrosis. miRNAs impact the expression of specific ligands, receptors, and signaling pathways, thus modulating EMT and consequently influencing fibrosis. This review summarizes the current knowledge concerning how miRNAs regulate EMT and highlights the specific roles that miRNAs-regulated EMT plays in fibrotic diseases as diverse as pulmonary fibrosis, hepatic fibrosis, renal fibrosis and cardiac fibrosis. It is desirable that a more comprehensive understanding of the functions of miRNAs-regulated EMT will facilitate the development of novel diagnostic and therapeutic strategies for various debilitating organ fibrosis.
Collapse
Affiliation(s)
- Xiao-Zhou Zou
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Ting Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China
| | - Zhi-Cheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chang-Ping Hu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan 410078, China.
| | - Zheng Zhang
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Central South University, Changsha, Hunan 410078, China.
| |
Collapse
|
46
|
Abstract
BACKGROUND The activation of the Notch signaling pathway has been shown to play an important role in diabetic nephropathy (DN) development. Besides, Notch-1 is a target gene in miR-34a. However, the regulation of the podocyte lesions involved in DN by miR-34a has not been identified. METHODS This study utilized miR-34a mimics and small interfering RNA transfection to construct miR-34a overexpression and lower-expression model to investigate the effect of miR-34a on the regulation of the Notch signaling pathway and podocyte lesions in DN. Western blotting and real-time quantitative polymerase chain reaction were applied for the quantitative testing of mRNA and protein expression. Apoptosis of podocyte was detected by TUNEL staining. RESULTS In high-glucose (HG) conditions, miR-34a overexpression inhibited the expression of Notch 1, Jagged 1, NICD, Hes 1, and Hey 1 proteins. Further, cleaved caspase-3, Bax, and phosphorylation of p53 (p-p53) were reduced significantly. Therefore, miR-34a overexpression inhibited the Notch signaling pathway and podocyte lesions induced by HG. β-arrestin was slightly reduced in HG conditions. Meanwhile, miR-34a overexpression could remit the inhibition. CONCLUSION Results from this study provide evidence that miR-34a may offer a new approach for the treatment of diabetes.
Collapse
Affiliation(s)
- Xiangying Zhang
- Department of Endocrinology, Tianjin Hospital, Tianjin, P.R. China
- Correspondence: Xiangying Zhang, Department of Endocrinology, Tianjin Hospital. Liberation South Road No. 406, Hexi District, Tianjin 300211, China (e-mail: )
| | | | | |
Collapse
|
47
|
Epigallocatechin-3-gallate attenuates cadmium-induced chronic renal injury and fibrosis. Food Chem Toxicol 2016; 96:70-8. [PMID: 27474435 DOI: 10.1016/j.fct.2016.07.030] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 07/13/2016] [Accepted: 07/25/2016] [Indexed: 12/11/2022]
Abstract
Cadmium (Cd) pollution is a serious environmental problem. Kidney is a main target organ of Cd toxicity. This study was undertaken to investigate the potential protective effects of epigallocatechin-3-gallate (EGCG) against chronic renal injury and fibrosis induced by CdCl2. Rat model was induced by exposing to 250 mg/L CdCl2 through drinking water. The renal function was evaluated by detecting the levels of blood urea nitrogen (BUN) and serum creatinine (SCR). The oxidative stress was measured by detecting the levels of malondialdehyde (MDA), nitric oxide (NO), reduced glutathione/oxidized glutathione (GSH/GSSG) and renal enzymatic antioxidant status. Additionally, the renal levels of transforming growth factor-β1 (TGF-β1), Smad3, phosphorylation-Smad3 (pp-Smad3), α-smooth muscle actin (α-SMA), vimentin and E-cadherin were measured by western blot assay. Renal levels of microRNA-21 (miR-21), miR-29a/b/c and miR-192 were measured by quantitative RT-PCR. It was found that EGCG ameliorated the CdCl2-induced renal injury, inhibited the level of oxidative stress, normalized renal enzymatic antioxidant status and E-cadherin level, as well as attenuated the over generation of TGF-β1, pp-Smad3, vimentin and α-SMA. EGCG also decreased the production of miR-21 and miR-192, and enhanced the levels of miR-29a/b/c. These results showed that EGCG could attenuate Cd induced chronic renal injury.
Collapse
|
48
|
Vaspin regulates the osteogenic differentiation of MC3T3-E1 through the PI3K-Akt/miR-34c loop. Sci Rep 2016; 6:25578. [PMID: 27156573 PMCID: PMC4860647 DOI: 10.1038/srep25578] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 04/18/2016] [Indexed: 01/15/2023] Open
Abstract
Vaspin (visceral adipose tissue-derived serine protease inhibitor) is a newly discovered adipokine that widely participates in diabetes mellitus, polycystic ovarian syndrome and other disorders of metabolism. However, the effect of vaspin on the regulation of osteogenesis and the mechanism responsible are still unclear. Here, we found that vaspin can attenuate the osteogenic differentiation of the preosteoblast cell line MC3T3-E1 in a dose-dependent way; also, during this process, the expression of miRNA-34c (miR-34c) was significantly increased. Down-regulation of the expression of miR-34c in MC3T3-E1 diminished the osteogenic inhibitory effect of vaspin, while the up-regulation of miR-34c increased this effect through its target gene Runx2. Meanwhile, we found that vaspin could also activate the PI3K-Akt signalling pathway. Blocking the PI3K-Akt signalling pathway with specific inhibitors could decrease the osteogenic inhibitory effect of vaspin as well as the expression level of miR-34c. Furthermore, knock-down of miR-34c could promote the activation of Akt, which was probably realised by targeting c-met expression. Thus, PI3K-Akt and miR-34c constituted a modulation loop and controlled the expression of each other. Taken together, our study showed that vaspin could inhibit the osteogenic differentiation in vitro, and the PI3K-Akt/miR-34c loop might be the underlying mechanism.
Collapse
|
49
|
Modeling the Transitions between Collective and Solitary Migration Phenotypes in Cancer Metastasis. Sci Rep 2015; 5:17379. [PMID: 26627083 PMCID: PMC4667179 DOI: 10.1038/srep17379] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 10/06/2015] [Indexed: 12/19/2022] Open
Abstract
Cellular plasticity during cancer metastasis is a major clinical challenge. Two key cellular plasticity mechanisms —Epithelial-to-Mesenchymal Transition (EMT) and Mesenchymal-to-Amoeboid Transition (MAT) – have been carefully investigated individually, yet a comprehensive understanding of their interconnections remains elusive. Previously, we have modeled the dynamics of the core regulatory circuits for both EMT (miR-200/ZEB/miR-34/SNAIL) and MAT (Rac1/RhoA). We now extend our previous work to study the coupling between these two core circuits by considering the two microRNAs (miR-200 and miR-34) as external signals to the core MAT circuit. We show that this coupled circuit enables four different stable steady states (phenotypes) that correspond to hybrid epithelial/mesenchymal (E/M), mesenchymal (M), amoeboid (A) and hybrid amoeboid/mesenchymal (A/M) phenotypes. Our model recapitulates the metastasis-suppressing role of the microRNAs even in the presence of EMT-inducing signals like Hepatocyte Growth Factor (HGF). It also enables mapping the microRNA levels to the transitions among various cell migration phenotypes. Finally, it offers a mechanistic understanding for the observed phenotypic transitions among different cell migration phenotypes, specifically the Collective-to-Amoeboid Transition (CAT).
Collapse
|
50
|
Sha WG, Shen L, Zhou L, Xu DY, Lu GY. Down-regulation of miR-186 contributes to podocytes apoptosis in membranous nephropathy. Biomed Pharmacother 2015; 75:179-84. [PMID: 26382839 DOI: 10.1016/j.biopha.2015.07.021] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 07/26/2015] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND AND AIM Podocytes apoptosis is the key process in the development of membranous nephropathy and miR-186 is reported to be related with cell apoptosis. Here we investigated the expression of miR-186 in membranous nephropathy (MGN) patients and the mechanism underlying the podocytes apoptosis. METHODS Thirty patients with MGN and 30 healthy people were included in this study. The expression of miR-186 was detected in renal tissue and podocyte cells exposed to AngII by real-time PCR. Caspase-3 activity was used to evaluate podocytes apoptosis. TLR4 and P2×7 protein expression was quantified by western blotting. miR-186 inhibitor and miR-186 mimic were transfected into cells to investigate the mechanism underlying miR-186 in podocytes apoptosis. RESULTS In MGN patients, the level of miR-186 was significantly down-regulated as well as the protein expression of TLR4 and P2×7 was up-regulated in renal tissue. In vitro experiments, TLR4 siRNA increased the expression of miR-186 and miR-186 inhibitor elevated the mRNA and protein expression of P2×7 in podocytes exposed to AngII. In addition, the level of cleaved-caspase-3 was up-regulated by miR-186 inhibitor. The TUNEL-positive cells and caspase-3 activity of podocytes induced by AngII were down-regulated by miR-186 mimic. CONCLUSIONS We revealed that TLR4 is involved in the regulation of miR-186 expression, and the anti-apoptotic effect of miR-186 on podocytes is correlated with P2×7 regulation.
Collapse
Affiliation(s)
- Wen-gang Sha
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Lei Shen
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Ling Zhou
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - De-yu Xu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China
| | - Guo-yuan Lu
- Department of Nephrology, The First Affiliated Hospital of Soochow University, Suzhou 215006, PR China.
| |
Collapse
|