1
|
Agwa SHA, Elzahwy SS, Hossam N, Yahia YA, Hamady S, Sherif N, Elshazly A, Darwish RM, Hashim JO, Adly MA, Abd Elsamee AM, Shamekh R, Roushdy MMS, Matboli M. Discriminatory power of a circulating multi-noncoding RNA panel in acute coronary syndrome subtypes: Towards precision detection. Int J Biochem Cell Biol 2024; 169:106531. [PMID: 38280541 DOI: 10.1016/j.biocel.2024.106531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/24/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
BACKGROUND Acute Coronary Syndrome (ACS) stands as a significant contributor to cardiovascular mortality, necessitating improved diagnostic tools for early detection and tailored therapeutic interventions. Current diagnostic modalities, exhibit limitations in sensitivity and specificity, urging the quest for novel biomarkers to enhance discrimination of the different stages of ACS including unstable angina, Non-ST-segment Elevation Myocardial Infarction (NSTEMI), and ST-segment Elevation Myocardial Infarction (STEMI). METHODS This study investigated the potential of a plasma-circulating multi-noncoding RNA (ncRNA) panel, comprising four miRNAs (miR-182-5p, miR-23a-3p, miR-146a-5p, and miR-183-5p) and three lncRNAs (SNHG15, SNHG5, and RMRP), selected based on their intricate involvement in ACS pathogenesis and signaling pathways regulating post-myocardial infarction (MI) processes. The differential expression of these ncRNAs was validated in sera of ACS patients and healthy controls via real-time polymerase chain reaction (RT-PCR). RESULTS Analysis revealed a marked upregulation of the multi-ncRNAs panel in ACS patients. Notably, miRNA-182-5p and lncRNA-RMRP exhibited exceptional discriminatory power, indicated by the high area under the curve (AUC) values (0.990 and 0.980, respectively). Importantly, this panel displayed superior efficacy in discriminating between STEMI and NSTEMI, outperforming conventional biomarkers like creatine kinase-MB and cardiac troponins. Additionally, the four miRNAs and lncRNA RMRP showcased remarkable proficiency in distinguishing between STEMI and unstable angina. CONCLUSION The findings underscore the promising potential of the multi-ncRNA panel as a robust tool for early ACS detection, and precise differentiation among ACS subtypes, and as a potential therapeutic target.
Collapse
Affiliation(s)
- Sara H A Agwa
- Clinical pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt.
| | - Sherif Samir Elzahwy
- Cardiovascular Medicine Department, Faculty of Medicine, Ain Shams University, Cairo 1382, Egypt
| | - Nourhan Hossam
- Medicinal Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Yahia A Yahia
- Biochemistry Department, Faculty of Pharmacy, Misr University for Science and Technology, Giza 12566, Egypt
| | - Shaimaa Hamady
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Nadine Sherif
- Clinical pathology and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Ahmed Elshazly
- Cardiovascular Medicine Department, Faculty of Medicine, Ain Shams University, Cairo 1382, Egypt
| | - Reham M Darwish
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Jomana Osama Hashim
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Mahmoud Ashraf Adly
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Aya M Abd Elsamee
- Biochemistry and Molecular Genomics Unit, Medical Ain Shams Research Institute (MASRI), Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Rania Shamekh
- Department of Pathology, University of South Florida, Tampa, FL 33620, USA
| | - Marian Maher Salib Roushdy
- Medicinal Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt
| | - Marwa Matboli
- Medicinal Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11382, Egypt.
| |
Collapse
|
2
|
El-Mancy SS, Boshra SA, Elnahas OS, Fayez SM, Sheta NM. Enhancement of Bottle Gourd Oil Activity via Optimized Self-Dispersing Lipid Formulations (SDLFs) to Mitigate Isoproterenol-Evoked Cardiac Toxicity in Rats via Modulating BMP, MMP2, and miRNA-21 and miRNA-23a Genes' Expression. Molecules 2023; 28:6168. [PMID: 37630419 PMCID: PMC10458851 DOI: 10.3390/molecules28166168] [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: 05/29/2023] [Revised: 08/17/2023] [Accepted: 08/19/2023] [Indexed: 08/27/2023] Open
Abstract
Bottle gourd (BG) oil (family Cucurbitaceae) has several pharmacological activities including a reduction of the hazard of cardiovascular and atherosclerosis conditions. This work aimed to develop and optimize self-dispersing lipid formulations (SDLFs) of BG oil by applying a full 32 factorial design. The formulation variables (oil concentration and surfactant mixture ratio) showed an obvious impact on the characters of the prepared BG-SDLFs including droplet size (DS), polydispersity index (PDI), emulsification time (ET), and transmission percentage (Tr%). The optimum BG-SDLF composed of 30% oil and Tween 80/Cremophor® RH40 (1:1) showed good emulsification characteristics and a better drug release profile compared with BG oil. In vivo study in isoproterenol-injected rats showed that BG oil and the optimized BG-SDLF improved cardiac function, by elevating the miRNA-23a gene expression level and decreasing miRNA-21 gene expression. They also caused the inhibition of the plasma B-type natriuretic peptide (BNP), N-terminal proatrial natriuretic peptide (NT-pro-BNP), cystatin c, galectin-3, lipoprotein-associated phospholipase A2 (Lp-PLA2), matrix metallopeptidase 2 (MMP2), cardiac troponin I (cTnI), and cardiac troponin T (cTnT). Our study demonstrated that BG oil and the optimized BG-SDLF provided a cardioprotection against isoproterenol-induced cardiac toxicity with better results in groups treated with the optimized BG-SDLF.
Collapse
Affiliation(s)
- Shereen S. El-Mancy
- Department of Pharmaceutics, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt; (S.S.E.-M.); (O.S.E.); (S.M.F.); (N.M.S.)
| | - Sylvia A. Boshra
- Department of Biochemistry, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt
| | - Osama S. Elnahas
- Department of Pharmaceutics, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt; (S.S.E.-M.); (O.S.E.); (S.M.F.); (N.M.S.)
| | - Sahar M. Fayez
- Department of Pharmaceutics, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt; (S.S.E.-M.); (O.S.E.); (S.M.F.); (N.M.S.)
| | - Nermin M. Sheta
- Department of Pharmaceutics, Faculty of Pharmacy, October 6 University, Giza 12585, Egypt; (S.S.E.-M.); (O.S.E.); (S.M.F.); (N.M.S.)
| |
Collapse
|
3
|
Turkieh A, El Masri Y, Pinet F, Dubois-Deruy E. Mitophagy Regulation Following Myocardial Infarction. Cells 2022; 11:cells11020199. [PMID: 35053316 PMCID: PMC8774240 DOI: 10.3390/cells11020199] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023] Open
Abstract
Mitophagy, which mediates the selective elimination of dysfunctional mitochondria, is essential for cardiac homeostasis. Mitophagy is regulated mainly by PTEN-induced putative kinase protein-1 (PINK1)/parkin pathway but also by FUN14 domain-containing 1 (FUNDC1) or Bcl2 interacting protein 3 (BNIP3) and BNIP3-like (BNIP3L/NIX) pathways. Several studies have shown that dysregulated mitophagy is involved in cardiac dysfunction induced by aging, aortic stenosis, myocardial infarction or diabetes. The cardioprotective role of mitophagy is well described, whereas excessive mitophagy could contribute to cell death and cardiac dysfunction. In this review, we summarize the mechanisms involved in the regulation of cardiac mitophagy and its role in physiological condition. We focused on cardiac mitophagy during and following myocardial infarction by highlighting the role and the regulation of PI NK1/parkin-; FUNDC1-; BNIP3- and BNIP3L/NIX-induced mitophagy during ischemia and reperfusion.
Collapse
|
4
|
Chen CC, Peng CC, Fan PC, Chu PH, Chang YS, Chang CH. Practical Procedures for Improving Detection of Circulating miRNAs in Cardiovascular Diseases. J Cardiovasc Transl Res 2020; 13:977-987. [PMID: 32440912 DOI: 10.1007/s12265-020-10019-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/28/2020] [Indexed: 10/24/2022]
Abstract
Hemolysis has been known to affect the measurement of circulating biomarkers. In this study, clinically applicable procedures for microRNA (miRNA) detection in serum samples of acute myocardial infarction patients were established. The 89 samples from patients admitted to the coronary care unit were collected. These samples obtained from heparin-treated and untreated patients were subjected to heparinase digestion prior to miRNA measurements by multiplex RT-qPCR. The good reproducibility of miRNA detection after heparinase digestion (average R2 = 0.97) indicated that this method can be used routinely for samples regardless of heparin medication. Additionally, the degree of hemolysis in these samples was highly related to the hemoglobin absorbance at 414 nm. Based on the hemoglobin absorbance, five hemolysis-associated miRNAs were identified in our data normalized with respect to both the spike-in control and the RNA amount in a given sample. Using these calibration procedures, miRNAs can be accurately quantified and identified for clinical samples. Graphical Abstract The practical procedures for miRNA detection in serum samples from the coronary care unit were established, and five hemolysis-associated miRNAs were accurately clarified through serial normalization.
Collapse
Affiliation(s)
- Chia-Chun Chen
- Molecular Medicine Research Center, Chang Gung University, No. 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 33302, Taiwan.
- Department of Colorectal Surgery, Chang Gung Memorial Hospital at Linkou, No. 5 Fusing Street, Guishan District, Taoyuan City, 333, Taiwan.
| | - Chen-Ching Peng
- Molecular Medicine Research Center, Chang Gung University, No. 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 33302, Taiwan
| | - Pei-Chun Fan
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, College of Medicine, Chang Gung University, No. 5 Fusing Street, Guishan District, Taoyuan City, 333, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, No. 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 33302, Taiwan
| | - Pao-Hsien Chu
- Department of Cardiology, Chang Gung Memorial Hospital, College of Medicine, Chang Gung University, 199 Tung Hwa North Road, Taipei, 105, Taiwan
| | - Yu-Sun Chang
- Molecular Medicine Research Center, Chang Gung University, No. 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 33302, Taiwan
- Graduate Institute of Biomedical Sciences, Chang Gung University, No. 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 33302, Taiwan
| | - Chih-Hsiang Chang
- Department of Nephrology, Kidney Research Center, Chang Gung Memorial Hospital, Linkou Medical Center, College of Medicine, Chang Gung University, No. 5 Fusing Street, Guishan District, Taoyuan City, 333, Taiwan
- Graduate Institute of Clinical Medical Sciences, Chang Gung University, No. 259 Wen-Hwa 1st Road, Kwei-Shan, Taoyuan, 33302, Taiwan
| |
Collapse
|
5
|
Yu FR, Xia YW, Wang SB, Xiao LH. Long noncoding RNA PVT1 facilitates high glucose-induced cardiomyocyte death through the miR-23a-3p/CASP10 axis. Cell Biol Int 2020; 45:154-163. [PMID: 33049089 DOI: 10.1002/cbin.11479] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 10/09/2020] [Accepted: 10/11/2020] [Indexed: 12/14/2022]
Abstract
Dilated cardiomyopathy (DCM) is the leading cause of morbidity and mortality in diabetic patients. Long noncoding RNA plasmacytoma variant translocation 1 (PVT1) has been shown to be related to the pathogenesis of DCM. However, the mechanism by which PVT1 regulates DCM pathogenesis is unclear. High glucose level was employed to construct a DCM cell model in vitro. Cell viability was determined via cell counting kit-8 assay. The level of lactate dehydrogenase (LDH) was measured with the corresponding kit. Expression levels of PVT1, miR-23a-3p, and caspase-10 (CASP10) messenger RNA were evaluated with a quantitative real-time polymerase chain reaction. Cell apoptosis was assessed by flow cytometry assay. Protein levels of B-cell lymphoma 2-associated X (Bax), cleaved-caspase-3 (cleaved-casp-3), and CASP10 were examined via western blot analysis. The relationship between PVT1 or CASP10 and miR-23a-3p was verified with dual-luciferase reporter assay. We observed that PVT1 and CASP10 were upregulated while miR-23a-3p was downregulated in high glucose-induced cardiomyocytes. High glucose levels repressed cardiomyocyte activity and induced cardiomyocyte apoptosis, but this influence was antagonized by PVT1 knockdown or miR-23a-3p overexpression. Furthermore, PVT1 acted as a sponge for miR-23a-3p, and miR-23a-3p inhibition counterbalanced the influence of PVT1 silencing on viability and apoptosis of cardiomyocytes under high glucose level treatment. PVT1 could increase CASP10 expression via sponging miR-23a-3p. In conclusion, PVT1 acted as a deleterious lncRNA in DCM. PVT1 facilitated cardiomyocyte death by regulating the miR-23a-3p/CASP10, which offered a new mechanism to comprehend the pathogenesis of DCM.
Collapse
Affiliation(s)
- Feng-Rong Yu
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| | - Yin-Wen Xia
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| | - Shao-Bo Wang
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| | - Li-Hua Xiao
- Department of Cardiology, Hanchuan City People's Hospital, Hanchuan, Hubei, China
| |
Collapse
|
6
|
Damasceno PKF, de Santana TA, Santos GC, Orge ID, Silva DN, Albuquerque JF, Golinelli G, Grisendi G, Pinelli M, Ribeiro Dos Santos R, Dominici M, Soares MBP. Genetic Engineering as a Strategy to Improve the Therapeutic Efficacy of Mesenchymal Stem/Stromal Cells in Regenerative Medicine. Front Cell Dev Biol 2020; 8:737. [PMID: 32974331 PMCID: PMC7471932 DOI: 10.3389/fcell.2020.00737] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem/stromal cells (MSCs) have been widely studied in the field of regenerative medicine for applications in the treatment of several disease settings. The therapeutic potential of MSCs has been evaluated in studies in vitro and in vivo, especially based on their anti-inflammatory and pro-regenerative action, through the secretion of soluble mediators. In many cases, however, insufficient engraftment and limited beneficial effects of MSCs indicate the need of approaches to enhance their survival, migration and therapeutic potential. Genetic engineering emerges as a means to induce the expression of different proteins and soluble factors with a wide range of applications, such as growth factors, cytokines, chemokines, transcription factors, enzymes and microRNAs. Distinct strategies have been applied to induce genetic modifications with the goal to enhance the potential of MCSs. This review aims to contribute to the update of the different genetically engineered tools employed for MSCs modification, as well as the factors investigated in different fields in which genetically engineered MSCs have been tested.
Collapse
Affiliation(s)
- Patricia Kauanna Fonseca Damasceno
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil
| | | | | | - Iasmim Diniz Orge
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil
| | - Daniela Nascimento Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil
| | | | - Giulia Golinelli
- Division of Oncology, Laboratory of Cellular Therapy, University of Modena and Reggio Emilia, Modena, Italy
| | - Giulia Grisendi
- Division of Oncology, Laboratory of Cellular Therapy, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Pinelli
- Division of Plastic Surgery, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Ricardo Ribeiro Dos Santos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil.,National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA), Rio de Janeiro, Brazil
| | - Massimo Dominici
- Division of Oncology, Laboratory of Cellular Therapy, University of Modena and Reggio Emilia, Modena, Italy
| | - Milena Botelho Pereira Soares
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Salvador, Brazil.,Health Institute of Technology, SENAI CIMATEC, Salvador, Brazil.,National Institute of Science and Technology for Regenerative Medicine (INCT-REGENERA), Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Park IH, Song YS, Joo HW, Shen GY, Seong JH, Shin NK, Cho YJ, Lee Y, Shin JH, Lim YH, Kim H, Kim KS. Role of MicroRNA-34a in Anti-Apoptotic Effects of Granulocyte-Colony Stimulating Factor in Diabetic Cardiomyopathy. Diabetes Metab J 2020; 44:173-185. [PMID: 31237127 PMCID: PMC7043984 DOI: 10.4093/dmj.2018.0211] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 01/14/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Recent studies have shown that microRNAs (miRNAs) are involved in the process of cardiomyocyte apoptosis. We have previously reported that granulocyte-colony stimulating factor (G-CSF) ameliorated diastolic dysfunction and attenuated cardiomyocyte apoptosis in a rat model of diabetic cardiomyopathy. In this study, we hypothesized a regulatory role of cardiac miRNAs in the mechanism of the anti-apoptotic effect of G-CSF in a diabetic cardiomyopathy rat model. METHODS Rats were given a high-fat diet and low-dose streptozotocin injection and then randomly allocated to receive treatment with either G-CSF or saline. H9c2 rat cardiomyocytes were cultured under a high glucose (HG) condition to induce diabetic cardiomyopathy in vitro. We examined the extent of apoptosis, miRNA expression, and miRNA target genes in the myocardium and H9c2 cells. RESULTS G-CSF treatment significantly decreased apoptosis and reduced miR-34a expression in diabetic myocardium and H9c2 cells under the HG condition. G-CSF treatment also significantly increased B-cell lymphoma 2 (Bcl-2) protein expression as a target for miR-34a. In addition, transfection with an miR-34a mimic significantly increased apoptosis and decreased Bcl-2 luciferase activity in H9c2 cells. CONCLUSION Our results indicate that G-CSF might have an anti-apoptotic effect through down-regulation of miR-34a in a diabetic cardiomyopathy rat model.
Collapse
Affiliation(s)
- In Hwa Park
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Yi Sun Song
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Hyun Woo Joo
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Guang Yin Shen
- Division of Cardiology, Department of Internal Medicine, Jilin Central Hospital, Jilin University, Jilin, China
| | - Jin Hee Seong
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Na Kyoung Shin
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
| | - Young Jong Cho
- Department of Laboratory Medicine, Kangwon National University School of Medicine, Chuncheon, Korea
| | - Yonggu Lee
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Jeong Hun Shin
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Young Hyo Lim
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea
| | - Hyuck Kim
- Department of Thoracic Surgery, Hanyang University Seoul Hospital, Seoul, Korea
| | - Kyung Soo Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, Korea
- Division of Cardiology, Department of Internal Medicine, Hanyang University College of Medicine, Seoul, Korea.
| |
Collapse
|
8
|
Zhao Z, Du S, Shen S, Wang L. microRNA‐132 inhibits cardiomyocyte apoptosis and myocardial remodeling in myocardial infarction by targeting IL‐1β. J Cell Physiol 2019; 235:2710-2721. [PMID: 31621911 DOI: 10.1002/jcp.29175] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/23/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Zonglei Zhao
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou China
| | - Song Du
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou China
| | - Shuxin Shen
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou China
| | - Lixia Wang
- Department of Cardiology Henan Provincial People's Hospital (Zhengzhou University People's Hospital) Zhengzhou China
| |
Collapse
|
9
|
Roufayel R, Kadry S. MicroRNAs: Crucial Regulators of Stress. Microrna 2019; 9:93-100. [PMID: 31241025 PMCID: PMC7366010 DOI: 10.2174/2211536608666190625120127] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 04/03/2019] [Accepted: 05/21/2019] [Indexed: 12/30/2022]
Abstract
Background
Signaling pathways including gene silencing, cellular differentiation, homeostasis, development and apoptosis are regulated and controlled by a wide range of miRNAs. Objective
Due to their potential binding sites in human-protein coding genes, many studies have also linked their altered expressions in various cancer types making them tumor suppressors agents. Methods
Moreover, each miRNA is predicted to have many mRNA targets indicating their extensive regulatory role in cell survival and developmental processes. Nowadays, diagnosis of early cancer stage development is now dependent on variable miRNA expression levels as potential oncogenic biomarkers in validating and targeting microRNAs for cancer therapy. Results
As the majority of miRNA, transcripts are derived from RNA polymerase II-directed transcription, stress response could result on a general reduction in the abundance of these transcripts. Over expression of various microRNAs have lead to B cell malignancy, potentiated KrasG12D-induced lung tumorigenesis, chronic lymphocytic leukemia, lymphoproliferative disease and autoimmunity. Conclusion
Altered miRNA expressions could have a significant impact on the abundance of proteins, making them attractive candidates as biomarkers for cancer detection and important regulators of apoptosis.
Collapse
Affiliation(s)
- Rabih Roufayel
- Department of Science, College of Engineering and Technology, American University of the Middle East, Egaila, Kuwait.,Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - Seifedine Kadry
- Department of Mathematics and Computer Science, Faculty of Science, Beirut Arab University, Beirut, Lebanon
| |
Collapse
|
10
|
Bukauskas T, Mickus R, Cereskevicius D, Macas A. Value of Serum miR-23a, miR-30d, and miR-146a Biomarkers in ST-Elevation Myocardial Infarction. Med Sci Monit 2019; 25:3925-3932. [PMID: 31130720 PMCID: PMC6556071 DOI: 10.12659/msm.913743] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The aim of this study was to analyze the relative expression level of miR-30d-5p, miR-23a-3p, and miR-146a-5p, and to comprehensively assess the diagnostic and predictive possibilities of these miRNAs. Their expression changes have not yet been sufficiently investigated during acute myocardial infarction. Therefore, it is important to comprehensively assess the diagnostic and predictive possibilities of these micro-ribonucleic acids (miRNAs). MATERIAL AND METHODS Random patients with ST‑elevated myocardial infarction (STEMI) were enrolled into the study group. The control group was comprised of patients with no inflammation or ischemic heart disease who were hospitalized for minor elective surgery. The relative expression level for each miRNA was determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR)-analysis. RESULTS There were 88 participants enrolled into the study: 62 patients were diagnosed with STEMI and there were 26 healthy controls. Expressions of miR-30d-5p, miR-146a-5p, and miR-23a-3p were respectively 1.581-fold, 4.048-fold, and 4.857-fold lower in patients with STEMI compared to the control group patients (all P values were <0.001). Downregulation of miR-23a-3p was significantly negatively correlated with risk scores of GRACE (Global Registry of Acute Coronary Events) and APACHE II (Acute Physiology and Chronic Health Evaluation II). MiR-23a-3p was a fair predictor for STEMI: area under the curve (AUC)=0.806. Cox regression analysis revealed that expression levels of analyzed miRNAs were not significantly associated with negative endpoints at 1 month after the onset of STEMI. CONCLUSIONS All investigated miRNAs were differentially expressed when comparing patients with STEMI and control group individuals. The evaluation of miR-23a-3p expression levels in serum could be useful to assess the severity of STEMI and as a potential diagnostic biomarker of this condition. In addition, miR-23a-3p may provide limited short-term prognostic value for STEMI patients.
Collapse
Affiliation(s)
- Tomas Bukauskas
- Department of Anesthesiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Rytis Mickus
- Medical Academy, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Darius Cereskevicius
- Department of Genetics and Molecular Medicine, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Andrius Macas
- Department of Anesthesiology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
11
|
Gong X, Liu H, Wang S, Liang S, Wang G. Exosomes derived from SDF1‐overexpressing mesenchymal stem cells inhibit ischemic myocardial cell apoptosis and promote cardiac endothelial microvascular regeneration in mice with myocardial infarction. J Cell Physiol 2019; 234:13878-13893. [PMID: 30720220 DOI: 10.1002/jcp.28070] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 12/14/2018] [Accepted: 12/18/2018] [Indexed: 01/23/2023]
Affiliation(s)
- Xu‐He Gong
- Department of Cardiology Beijing Friendship Hospital, Capital Medical University Beijing China
| | - Hui Liu
- Department of Cardiology State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| | - Si‐Jia Wang
- Department of Emergency Beijing Friendship Hospital, Capital Medical University Beijing China
| | - Si‐Wen Liang
- Department of Cardiology Beijing Friendship Hospital, Capital Medical University Beijing China
| | - Guo‐Gan Wang
- Department of Cardiology State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
| |
Collapse
|
12
|
Functionally Improved Mesenchymal Stem Cells to Better Treat Myocardial Infarction. Stem Cells Int 2018; 2018:7045245. [PMID: 30622568 PMCID: PMC6286742 DOI: 10.1155/2018/7045245] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/10/2018] [Accepted: 09/30/2018] [Indexed: 12/14/2022] Open
Abstract
Myocardial infarction (MI) is one of the leading causes of death worldwide. Mesenchymal stem cell (MSC) transplantation is considered a promising approach and has made significant progress in preclinical studies and clinical trials for treating MI. However, hurdles including poor survival, retention, homing, and differentiation capacity largely limit the therapeutic effect of transplanted MSCs. Many strategies such as preconditioning, genetic modification, cotransplantation with bioactive factors, and tissue engineering were developed to improve the survival and function of MSCs. On the other hand, optimizing the hostile transplantation microenvironment of the host myocardium is also of importance. Here, we review the modifications of MSCs as well as the host myocardium to improve the efficacy of MSC-based therapy against MI.
Collapse
|
13
|
Li S, Ren J, Sun Q. The expression of microRNA-23a regulates acute myocardial infarction in patients and in vitro through targeting PTEN. Mol Med Rep 2018; 17:6866-6872. [PMID: 29488607 DOI: 10.3892/mmr.2018.8640] [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: 09/11/2017] [Indexed: 11/05/2022] Open
Abstract
Cardiovascular disease is responsible for one of the highest rates of fatality worldwide. The present study investigated the presence and influence of microRNA (miRNA)-23a in the regulation of acute myocardial infarction (AMI). A total of 6 patients with AMI and 6 normal volunteers without myocardial disease were included, and blood samples were taken to analyze the expression of miRNA‑23a by reverse transcription‑quantitative polymerase chain reaction. miRNA‑23a expression in patients with AMI was downregulated compared with the normal group. In H9C2 cells treated with H2O2, upregulation of miRNA‑23a expression increased the superoxide dismutase, glutathione and catalase activity levels, and suppressed the malonaldehyde activity level, as determined by ELISA. Western blot analysis and a caspase‑3 substrate assay demonstrated that upregulation of miRNA‑23a expression suppressed the Bcl‑2‑associated X (Bax)/Bcl‑2 protein expression ratio, caspase‑3 activity level and tumor suppressor p53 (p53) protein expression in H2O2‑induced H9C2 cells. Furthermore, downregulation of phosphatase and tensin homolog (PTEN), by the PTEN inhibitor bpV(HOpic), increased miRNA‑23a expression and suppressed the Bax/Bcl‑2 protein expression ratio, caspase‑3 activity level and p53 protein expression in H2O2‑induced H9C2 cells. Therefore, the results of the present study indicate that the expression of miRNA‑23a may regulate AMI through targeting PTEN in patients and in vitro, and PTEN/miRNA‑23a may therefore be potential targets for the clinical treatment of AMI.
Collapse
Affiliation(s)
- Shengli Li
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100022, P.R. China
| | - Jie Ren
- Department of Medical Cardiology, The First Affiliated Hospital of Xi'an Jiao Tong University, Xi'an, Shaanxi 710061, P.R. China
| | - Qianmei Sun
- Department of Internal Medicine, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100022, P.R. China
| |
Collapse
|
14
|
Wu R, Hu X, Wang J. Concise Review: Optimized Strategies for Stem Cell-Based Therapy in Myocardial Repair: Clinical Translatability and Potential Limitation. Stem Cells 2018; 36:482-500. [PMID: 29330880 DOI: 10.1002/stem.2778] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 12/28/2017] [Accepted: 12/31/2017] [Indexed: 12/15/2022]
Abstract
Ischemic heart diseases (IHDs) remain major public health problems with high rates of morbidity and mortality worldwide. Despite significant advances, current therapeutic approaches are unable to rescue the extensive and irreversible loss of cardiomyocytes caused by severe ischemia. Over the past 16 years, stem cell-based therapy has been recognized as an innovative strategy for cardiac repair/regeneration and functional recovery after IHDs. Although substantial preclinical animal studies using a variety of stem/progenitor cells have shown promising results, there is a tremendous degree of skepticism in the clinical community as many stem cell trials do not confer any beneficial effects. How to accelerate stem cell-based therapy toward successful clinical application attracts considerate attention. However, many important issues need to be fully addressed. In this Review, we have described and compared the effects of different types of stem cells with their dose, delivery routes, and timing that have been routinely tested in recent preclinical and clinical findings. We have also discussed the potential mechanisms of action of stem cells, and explored the role and underlying regulatory components of stem cell-derived secretomes/exosomes in myocardial repair. Furthermore, we have critically reviewed the different strategies for optimizing both donor stem cells and the target cardiac microenvironments to enhance the engraftment and efficacy of stem cells, highlighting their clinical translatability and potential limitation. Stem Cells 2018;36:482-500.
Collapse
Affiliation(s)
- Rongrong Wu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Xinyang Hu
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| | - Jian'an Wang
- Department of Cardiology, Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, People's Republic of China.,Cardiovascular Key Laboratory of Zhejiang Province, Hangzhou, People's Republic of China
| |
Collapse
|
15
|
Miao C, Lei M, Hu W, Han S, Wang Q. A brief review: the therapeutic potential of bone marrow mesenchymal stem cells in myocardial infarction. Stem Cell Res Ther 2017; 8:242. [PMID: 29096705 PMCID: PMC5667518 DOI: 10.1186/s13287-017-0697-9] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Myocardial infarction (MI) results in dysfunction and irreversible loss of cardiomyocytes and is among the most serious health threats today. Bone marrow mesenchymal stem cells (BMSCs), with their capacity for multidirectional differentiation, low immunogenicity, and high portability, can serve as ideal seed cells in cardiovascular disease therapy. In this review, we examine recent literature concerning the application of BMSCs for the treatment of MI and consider the following aspects: activity of transplanted cells, migration and homing of BMSCs, immunomodulatory and anti-inflammatory effects of BMSCs, anti-fibrotic activity of BMSCs, the role of BMSCs in angiogenesis, and differentiation of BMSCs into cardiomyocyte-like cells and endothelial cells. Each aspect is complementary to the others and together they promote the repair of cardiomyocytes by BMSCs after MI. Although transplantation of BMSCs has enabled new options for MI treatment, the critical issue we must now address is the reduced viability of transplanted BMSCs due to inadequate blood supply, poor nourishment of cells, and generation of free radicals. More clinical trials are needed to prove the therapeutic potential of BMSCs in MI.
Collapse
Affiliation(s)
- Chi Miao
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Mingming Lei
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Weina Hu
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Shuo Han
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China
| | - Qi Wang
- Cardiovascular Department Of Internal Medicine, The Fourth Affiliated Hospital of China Medical University, Chongshandong Street No.4, Shenyang, 110032, China.
| |
Collapse
|
16
|
Roufayel R, Kadry S. Expression of miR-23a by apoptotic regulators in human cancer: A review. Cancer Biol Ther 2017; 18:269-276. [PMID: 28453394 DOI: 10.1080/15384047.2017.1310342] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
MicroRNAs play fundamental roles in mammalian development, differentiation and cellular homeostasis by regulating essential processes such as proliferation, migration, metabolism, migration and cell death. These small non-coding RNAs are also responsible in RNA silencing, and in many developmental and pathological processes. Not surprisingly, miR-23a misexpression contributes to numerous diseases including cancer where certain miRNA genes have been classified as either oncogenes or tumor suppressor genes. Since a single microRNA is capable of targeting a large number of mRNA sequences, de-regulated miRNA expression has the ability to alter various transcripts and activate a wide range of cancer-related pathways. This review article documents reduced levels of mature miR-23a in various tumors, primarily due to epigenetic silencing or alterations in biogenesis pathways. Moreover, inhibition of miR-23a in stressed cells represent a general mechanism for inducing apoptosis and these microRNAs are showed to be regulated by molecular chaperon HSP70. Microarray expression analysis of miRNA overexpression or depletion is now used in the characterization of cancer development pathways and as a biomarker for early cancer detection.
Collapse
Affiliation(s)
- Rabih Roufayel
- a Department of Science , American University of the Middle East , Kuwait
| | - Seifedine Kadry
- a Department of Science , American University of the Middle East , Kuwait
| |
Collapse
|
17
|
Verjans R, van Bilsen M, Schroen B. MiRNA Deregulation in Cardiac Aging and Associated Disorders. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 334:207-263. [PMID: 28838539 DOI: 10.1016/bs.ircmb.2017.03.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The prevalence of age-related diseases is increasing dramatically, among which cardiac disease represents the leading cause of death. Aging of the heart is characterized by various molecular and cellular hallmarks impairing both cardiomyocytes and noncardiomyocytes, and resulting in functional deteriorations of the cardiac system. The aging process includes desensitization of β-adrenergic receptor (βAR)-signaling and decreased calcium handling, altered growth signaling and cardiac hypertrophy, mitochondrial dysfunction and impaired autophagy, increased programmed cell death, low-grade inflammation of noncanonical inflammatory cells, and increased ECM deposition. MiRNAs play a fundamental role in regulating the processes underlying these detrimental changes in the cardiac system, indicating that MiRNAs are crucially involved in aging. Among others, MiR-34, MiR-146a, and members of the MiR-17-92 cluster, are deregulated during senescence and drive cardiac aging processes. It is therefore suggested that MiRNAs form possible therapeutic targets to stabilize the aged failing myocardium.
Collapse
Affiliation(s)
- Robin Verjans
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Marc van Bilsen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Blanche Schroen
- Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
18
|
miR clusters target cellular functional complexes by defining their degree of regulatory freedom. Cancer Metastasis Rev 2017; 35:289-322. [PMID: 26970968 DOI: 10.1007/s10555-016-9617-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Using the two paralog miR-23∼27∼24 clusters as an example and combining experimental and clinical data in a systematical approach to microRNA (miR) function and dysregulation, a complex picture of their roles in cancer is drawn. Various findings appear to be contradictory to a larger extent and cannot be fully explained by the classical regulatory network models and feedback loops that are mainly considered by one-to-one regulatory interactions of the involved molecules. Here, we propose an extended model of the regulatory role of miRs that, at least, supplements the usually considered single/oligo-target regulation of certain miRs. The cellular availability of the participating miR members in this model reflects an upper hierarchy level of intracellular and extracellular environmental influences, such as neighboring cells, soluble factors, hypoxia, chemotherapeutic drugs, and irradiation, among others. The novel model is based on the understanding of cellular functional complexes, such as for apoptosis, migration, and proliferation. These complexes consist of many regulatory components that can be targeted by miR cluster members to a different extent but may affect the functional complex in different ways. We propose that the final miR-related effect is a result of the possible degree of regulatory freedom provided by the miR effects on the whole functional complex structure. This degree of regulatory freedom defines to which extent the cellular functional complex can react in response to regulatory triggers, also understood as sensitization (more regulatory response options) or de-sensitization (less regulatory response options) of the system rather than single molecules.
Collapse
|
19
|
Karpov AA, Udalova DV, Pliss MG, Galagudza MM. Can the outcomes of mesenchymal stem cell-based therapy for myocardial infarction be improved? Providing weapons and armour to cells. Cell Prolif 2016; 50. [PMID: 27878916 DOI: 10.1111/cpr.12316] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 10/12/2016] [Indexed: 02/06/2023] Open
Abstract
Use of mesenchymal stem cell (MSC) transplantation after myocardial infarction (MI) has been found to have infarct-limiting effects in numerous experimental and clinical studies. However, recent meta-analyses of randomized clinical trials on MSC-based MI therapy have highlighted the need for improving its efficacy. There are two principal approaches for increasing therapeutic effect of MSCs: (i) preventing massive MSC death in ischaemic tissue and (ii) increasing production of cardioreparative growth factors and cytokines with transplanted MSCs. In this review, we aim to integrate our current understanding of genetic approaches that are used for modification of MSCs to enable their improved survival, engraftment, integration, proliferation and differentiation in the ischaemic heart. Genetic modification of MSCs resulting in increased secretion of paracrine factors has also been discussed. In addition, data on MSC preconditioning with physical, chemical and pharmacological factors prior to transplantation are summarized. MSC seeding on three-dimensional polymeric scaffolds facilitates formation of both intercellular connections and contacts between cells and the extracellular matrix, thereby enhancing cell viability and function. Use of genetic and non-genetic approaches to modify MSC function holds great promise for regenerative therapy of myocardial ischaemic injury.
Collapse
Affiliation(s)
- Andrey A Karpov
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia.,Department of Pathophysiology, First Pavlov State Medical University of Saint Petersburg, St Petersburg, Russia
| | - Daria V Udalova
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia
| | - Michael G Pliss
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia
| | - Michael M Galagudza
- Institute of Experimental Medicine, Federal Almazov North-West Medical Research Centre, St Petersburg, Russia.,ITMO University, St Petersburg, Russia
| |
Collapse
|
20
|
Trindade F, Leite-Moreira A, Ferreira-Martins J, Ferreira R, Falcão-Pires I, Vitorino R. Towards the standardization of stem cell therapy studies for ischemic heart diseases: Bridging the gap between animal models and the clinical setting. Int J Cardiol 2016; 228:465-480. [PMID: 27870978 DOI: 10.1016/j.ijcard.2016.11.236] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 12/20/2022]
Abstract
Today there is an increasing demand for heart transplantations for patients diagnosed with heart failure. Though, shortage of donors as well as the large number of ineligible patients hurdle such treatment option. This, in addition to the considerable number of transplant rejections, has driven the clinical research towards the field of regenerative medicine. Nonetheless, to date, several stem cell therapies tested in animal models fall by the wayside and when they meet the criteria to clinical trials, subjects often exhibit modest improvements. A main issue slowing down the admission of such therapies in the domain of human trials is the lack of protocol standardization between research groups, which hampers comparison between different approaches as well as the lack of thought regarding the clinical translation. In this sense, given the large amount of reports on stem cell therapy studies in animal models reported in the last 3years, we sought to evaluate their advantages and limitations towards the clinical setting and provide some suggestions for the forthcoming investigations. We expect, with this review, to start a new paradigm on regenerative medicine, by evoking the debate on how to plan novel stem cell therapy studies with animal models in order to achieve more consistent scientific production and accelerate the admission of stem cell therapies in the clinical setting.
Collapse
Affiliation(s)
- Fábio Trindade
- iBiMED, Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Portugal.
| | - Adelino Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Portugal
| | | | - Rita Ferreira
- QOPNA, Mass Spectrometry Center, Department of Chemistry, University of Aveiro, Portugal
| | - Inês Falcão-Pires
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Portugal
| | - Rui Vitorino
- iBiMED, Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Portugal; Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Portugal.
| |
Collapse
|
21
|
Bobis-Wozowicz S, Kmiotek K, Kania K, Karnas E, Labedz-Maslowska A, Sekula M, Kedracka-Krok S, Kolcz J, Boruczkowski D, Madeja Z, Zuba-Surma EK. Diverse impact of xeno-free conditions on biological and regenerative properties of hUC-MSCs and their extracellular vesicles. J Mol Med (Berl) 2016; 95:205-220. [PMID: 27638341 PMCID: PMC5239805 DOI: 10.1007/s00109-016-1471-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/30/2016] [Accepted: 09/01/2016] [Indexed: 12/13/2022]
Abstract
Growing evidence indicates that intracellular signaling mediated by extracellular vesicles (EVs) released by stem cells plays a considerable role in triggering the regenerative program upon transplantation. EVs from umbilical cord mesenchymal stem cells (UC-MSC-EVs) have been shown to enhance tissue repair in animal models. However, translating such results into clinical practice requires optimized EV collection procedures devoid of animal-originating agents. Thus, in this study, we analyzed the influence of xeno-free expansion media on biological properties of UC-MSCs and UC-MSC-EVs for future applications in cardiac repair in humans. Our results show that proliferation, differentiation, phenotype stability, and cytokine secretion by UC-MSCs vary depending on the type of xeno-free media. Importantly, we found distinct molecular and functional properties of xeno-free UC-MSC-EVs including enhanced cardiomyogenic and angiogenic potential impacting on target cells, which may be explained by elevated concentration of several pro-cardiogenic and pro-angiogenic microRNA (miRNAs) present in the EVs. Our data also suggest predominantly low immunogenic capacity of certain xeno-free UC-MSC-EVs reflected by their inhibitory effect on proliferation of immune cells in vitro. Summarizing, conscious selection of cell culture conditions is required to harvest UC-MSC-EVs with the optimal desired properties including enhanced cardiac and angiogenic capacity, suitable for tissue regeneration. KEY MESSAGE Type of xeno-free media influences biological properties of UC-MSCs in vitro. Certain xeno-free media promote proliferation and differentiation ability of UC-MSCs. EVs collected from xeno-free cultures of UC-MSCs are biologically active. Xeno-free UC-MSC-EVs enhance cardiac and angiogenic potential of target cells. Type of xeno-free media determines immunomodulatory effects mediated by UC-MSC-EVs.
Collapse
Affiliation(s)
- Sylwia Bobis-Wozowicz
- Department of Cell Biology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.
| | - Katarzyna Kmiotek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Karolina Kania
- Department of Cell Biology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Elzbieta Karnas
- Department of Cell Biology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.,Malopolska Centre of Biotechnology, 30-387, Krakow, Poland
| | - Anna Labedz-Maslowska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | | | - Sylwia Kedracka-Krok
- Department of Physical Biochemistry, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Jacek Kolcz
- Department of Pediatric Cardiac Surgery, Polish-American Children's Hospital, 30-663, Krakow, Poland
| | | | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland
| | - Ewa K Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics, and Biotechnology, Jagiellonian University, 30-387, Krakow, Poland.
| |
Collapse
|
22
|
Abstract
Although cardiac resuscitation can revive the whole body, the mechanisms are unclear. To this end, we propose that reviving a dead/dysfunctional cardiomyocyte will shed light on resuscitation mechanisms and pave the way to treat cardiac myopathies. The degradation of the myocyte cytoskeleton by the proteasome system which involves calpains, ubiquitin, caspases and matrix metalloproteases is the main focus of this review. The activation of calpains beyond the calpastatin-mediated inhibition due to extensive calcium harbor can lead to titin degradation, damage to the sarcomere and contractile dysfunction. The ubiquitin proteasome system can disturb the protein homeostasis within the cell and generate a dysfunctional myocyte. The matrix metalloproteases disrupt the collagen/elastin ratio and connexins to generate arrhythmias. The concept of cardiac resuscitation stems from protecting the myocyte cytoskeleton and keeping the protein homeostasis intact through management of the degradation machinery. In this regard, proteasome inhibitors for the degradation machinery have an elegant space. Recently exosomes have been identified potentially, as carriers of microRNAs or proteins that can modify the target cells. Exosomes loaded with the inhibitor "cargo" which comprises microRNAs, siRNAs or proteins to inhibit the degradation machinery can be a method of choice for cardiac resuscitation-a process difficult to execute.
Collapse
|
23
|
Singh A, Singh A, Sen D. Mesenchymal stem cells in cardiac regeneration: a detailed progress report of the last 6 years (2010-2015). Stem Cell Res Ther 2016; 7:82. [PMID: 27259550 PMCID: PMC4893234 DOI: 10.1186/s13287-016-0341-0] [Citation(s) in RCA: 149] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Mesenchymal stem cells have been used for cardiovascular regenerative therapy for decades. These cells have been established as one of the potential therapeutic agents, following several tests in animal models and clinical trials. In the process, various sources of mesenchymal stem cells have been identified which help in cardiac regeneration by either revitalizing the cardiac stem cells or revascularizing the arteries and veins of the heart. Although mesenchymal cell therapy has achieved considerable admiration, some challenges still remain that need to be overcome in order to establish it as a successful technique. This in-depth review is an attempt to summarize the major sources of mesenchymal stem cells involved in myocardial regeneration, the significant mechanisms involved in the process with a focus on studies (human and animal) conducted in the last 6 years and the challenges that remain to be addressed.
Collapse
Affiliation(s)
- Aastha Singh
- School of Bio Sciences and Technology, VIT University, Vellore, India
| | - Abhishek Singh
- School of Bio Sciences and Technology, VIT University, Vellore, India
| | - Dwaipayan Sen
- School of Bio Sciences and Technology, VIT University, Vellore, India. .,Cellular and Molecular Therapeutics Laboratory, Centre for Biomaterials, Cellular and Molecular Theranostics (CBCMT), VIT University, Vellore, 632014, Tamil Nadu, India.
| |
Collapse
|
24
|
Wei B, Bai X, Chen K, Zhang X. SP600125 enhances the anti-apoptotic capacity and migration of bone marrow mesenchymal stem cells treated with tumor necrosis factor-α. Biochem Biophys Res Commun 2016; 475:301-7. [PMID: 27233606 DOI: 10.1016/j.bbrc.2016.05.107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 05/21/2016] [Indexed: 01/21/2023]
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are chronic disorders associated with inflammation of joints characterized by damage to the underlying cartilage and bone. Bone marrow mesenchymal stem cells (BMSCs) are candidates for regeneration of bone and cartilage, which is inhibited by inflammatory cytokines in OA and RA, in particular tumor necrosis factor-α (TNF-α). This study aimed to investigate if the c-Jun N-terminal kinases (JNK)-specific inhibitor SP600125 could enhance the anti-apoptosis and migration of BMSCs treated with TNF-α. The level of apoptosis was evaluated via terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)/4',6-diamidino-2-phenylindole (DAPI) staining, annexin V/propidium iodide (PI) staining and western blotting. Migration of BMSCs was assessed using transwell migration chambers. We showed that the survival capacity and migration of BMSCs was significantly inhibited by TNF-α, which was blocked by pretreatment with SP600125. In the presence of SP600125, expression of cleaved caspase-9/-3 and p53 as well as the ratio of Bax to Bcl-2 was significantly decreased compared to treatment with TNF-α alone. Our results therefore indicate that SP600125 improves the migration capacity of TNF-α-treated BMSCs and exerts a significant effect on the viability of TNF-α-treated BMSCs through reducing the up-regulation of p53, caspase-9/-3 and the Bcl-2 family induced by TNF-α. These findings suggest that SP600125 is of potential use in promoting the regeneration of bone and cartilage in OA and RA.
Collapse
Affiliation(s)
- Bo Wei
- Department of Orthopedics, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China; Department of Sports Medicine and Joint Surgery, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China
| | - Xizhuang Bai
- Department of Orthopedics, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China; Department of Sports Medicine and Joint Surgery, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China.
| | - Kang Chen
- Department of Orthopedics, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China; Department of Sports Medicine and Joint Surgery, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China
| | - Xiaonan Zhang
- Department of Orthopedics, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China; Department of Sports Medicine and Joint Surgery, The People's Hospital of China Medical University, Shenhe, Shenyang, Liaoning 110016, China
| |
Collapse
|
25
|
Nollet E, Hoymans VY, Van Craenenbroeck AH, Vrints CJ, Van Craenenbroeck EM. Improving stem cell therapy in cardiovascular diseases: the potential role of microRNA. Am J Physiol Heart Circ Physiol 2016; 311:H207-18. [PMID: 27208159 DOI: 10.1152/ajpheart.00239.2016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/11/2016] [Indexed: 11/22/2022]
Abstract
The initial promising prospect of autologous bone marrow-derived stem cell therapy in the setting of cardiovascular diseases has been overshadowed by functional shortcomings of the stem cell product. As powerful epigenetic regulators of (stem) cell function, microRNAs are valuable targets for novel therapeutic strategies. Indeed, modulation of specific miRNA expression could contribute to improved therapeutic efficacy of stem cell therapy. First, this review elaborates on the functional relevance of miRNA dysregulation in bone marrow-derived progenitor cells in different cardiovascular diseases. Next, we provide a comprehensive overview of the current evidence on the effect of specific miRNA modulation in several types of progenitor cells on cardiac and/or vascular regeneration. By elaborating on the cardioprotective regulation of progenitor cells on cardiac miRNAs, more insight in the underlying mechanisms of stem cell therapy is provided. Finally, some considerations are made regarding the potential of circulating miRNAs as regulators of the miRNA signature of progenitor cells in cardiovascular diseases.
Collapse
Affiliation(s)
- Evelien Nollet
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
| | - Vicky Y Hoymans
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium
| | - Amaryllis H Van Craenenbroeck
- Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium; Department of Nephrology, Antwerp University Hospital, Antwerp, Belgium; Laboratory of Experimental Medicine and Pediatrics, University of Antwerp, Antwerp, Belgium; and
| | - Christiaan J Vrints
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- Laboratory of Cellular and Molecular Cardiology, Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium; Cardiovascular Diseases, Department of Translational Pathophysiological Research, University of Antwerp, Antwerp, Belgium; Department of Cardiology, Antwerp University Hospital, Antwerp, Belgium
| |
Collapse
|
26
|
Yang R, Ouyang Y, Li W, Wang P, Deng H, Song B, Hou J, Chen Z, Xie Z, Liu Z, Li J, Cen S, Wu Y, Shen H. Autophagy Plays a Protective Role in Tumor Necrosis Factor-α-Induced Apoptosis of Bone Marrow-Derived Mesenchymal Stem Cells. Stem Cells Dev 2016; 25:788-97. [PMID: 26985709 DOI: 10.1089/scd.2015.0387] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Bone marrow-derived mesenchymal stem cells (BMSCs) are being broadly investigated for treating numerous inflammatory diseases. However, the low survival rate of BMSCs during the transplantation process has limited their application. Autophagy can maintain cellular homeostasis and protect cells against environmental stresses. Tumor necrosis factor-α (TNF-α) is an important inflammatory cytokine that can induce both autophagy and apoptosis of BMSCs. However, the actual role of autophagy in TNF-α-induced apoptosis of BMSCs remains poorly understood. In the current study, BMSCs were treated with TNF-α/cycloheximide (CHX), and cell death was examined by the Cell Counting Kit-8, Hoechst 33342 staining, and flow cytometric analysis as well as by the level of caspase-3 and caspase-8. Meanwhile, autophagic flux was examined by analyzing the level of microtubule-associated protein light chain 3 B (LC3B)-II and SQSTEM1/p62 and by examining the amount of green fluorescent protein-LC3B by fluorescence microscopy. Then, the cell death and autophagic flux of BMSCs were examined after pretreatment and cotreatment with 3-methyladenine (3-MA, autophagy inhibitor) or rapamycin (Rap, autophagy activator) together with TNF-α/CHX. Moreover, BMSCs pretreated with lentiviruses encoding short hairpin RNA of beclin-1 (BECN1) were treated with TNF-α/CHX, and then cell death and autophagic flux were detected. We showed that BMSCs treated with TNF-α/CHX presented dramatically elevated autophagic flux and cell death. Furthermore, we showed that 3-MA and shBECN1 treatment accelerated TNF-α/CHX-induced apoptosis, but that Rap treatment ameliorated cell death. Our results demonstrate that autophagy protects BMSCs against TNF-α-induced apoptosis. Enhancing the autophagy of BMSCs may elevate cellular survival in an inflammatory microenvironment.
Collapse
Affiliation(s)
- Rui Yang
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Yi Ouyang
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Weiping Li
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Peng Wang
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Haiquan Deng
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Bin Song
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Jingyi Hou
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Zhong Chen
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Zhongyu Xie
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Zhenhua Liu
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Jinteng Li
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Shuizhong Cen
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Yanfeng Wu
- 2 Center for Biotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| | - Huiyong Shen
- 1 Department of Orthopedics, Sun Yat-sen Memorial Hospital, Sun Yat-sen University , Guangzhou, People's Republic of China
| |
Collapse
|
27
|
Abdelwahid E, Kalvelyte A, Stulpinas A, de Carvalho KAT, Guarita-Souza LC, Foldes G. Stem cell death and survival in heart regeneration and repair. Apoptosis 2016; 21:252-68. [PMID: 26687129 PMCID: PMC5200890 DOI: 10.1007/s10495-015-1203-4] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cardiovascular diseases are major causes of mortality and morbidity. Cardiomyocyte apoptosis disrupts cardiac function and leads to cardiac decompensation and terminal heart failure. Delineating the regulatory signaling pathways that orchestrate cell survival in the heart has significant therapeutic implications. Cardiac tissue has limited capacity to regenerate and repair. Stem cell therapy is a successful approach for repairing and regenerating ischemic cardiac tissue; however, transplanted cells display very high death percentage, a problem that affects success of tissue regeneration. Stem cells display multipotency or pluripotency and undergo self-renewal, however these events are negatively influenced by upregulation of cell death machinery that induces the significant decrease in survival and differentiation signals upon cardiovascular injury. While efforts to identify cell types and molecular pathways that promote cardiac tissue regeneration have been productive, studies that focus on blocking the extensive cell death after transplantation are limited. The control of cell death includes multiple networks rather than one crucial pathway, which underlies the challenge of identifying the interaction between various cellular and biochemical components. This review is aimed at exploiting the molecular mechanisms by which stem cells resist death signals to develop into mature and healthy cardiac cells. Specifically, we focus on a number of factors that control death and survival of stem cells upon transplantation and ultimately affect cardiac regeneration. We also discuss potential survival enhancing strategies and how they could be meaningful in the design of targeted therapies that improve cardiac function.
Collapse
Affiliation(s)
- Eltyeb Abdelwahid
- Feinberg School of Medicine, Feinberg Cardiovascular Research Institute, Northwestern University, 303 E. Chicago Ave., Tarry 14-725, Chicago, IL, 60611, USA.
| | - Audrone Kalvelyte
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Aurimas Stulpinas
- Department of Molecular Cell Biology, Vilnius University Institute of Biochemistry, Vilnius, Lithuania
| | - Katherine Athayde Teixeira de Carvalho
- Cell Therapy and Biotechnology in Regenerative Medicine Research Group, Pequeno Príncipe Faculty, Pelé Pequeno Príncipe Institute, Curitiba, Paraná, 80250-200, Brazil
| | - Luiz Cesar Guarita-Souza
- Experimental Laboratory of Institute of Biological and Health Sciences of Pontifical Catholic University of Parana, Curitiba, Paraná, 80215-901, Brazil
| | - Gabor Foldes
- National Heart and Lung Institute, Imperial College London, Imperial Centre for Experimental and Translational Medicine, Du Cane Road, London, W12 0NN, UK
| |
Collapse
|
28
|
The role of microRNAs in coronary artery disease: From pathophysiology to diagnosis and treatment. Atherosclerosis 2015; 241:624-33. [PMID: 26117399 DOI: 10.1016/j.atherosclerosis.2015.06.037] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 05/31/2015] [Accepted: 06/17/2015] [Indexed: 01/08/2023]
|
29
|
Morey TM, Roufayel R, Johnston DS, Fletcher AS, Mosser DD. Heat shock inhibition of CDK5 increases NOXA levels through miR-23a repression. J Biol Chem 2015; 290:11443-54. [PMID: 25829494 DOI: 10.1074/jbc.m114.625988] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Indexed: 11/06/2022] Open
Abstract
Hyperthermia is a proteotoxic stress that is lethal when exposure is extreme but also cytoprotective in that sublethal exposure leads to the synthesis of heat shock proteins, including HSP70, which are able to inhibit stress-induced apoptosis. CDK5 is an atypical cyclin-dependent kinase family member that regulates many cellular functions including motility and survival. Here we show that exposure of a human lymphoid cell line to hyperthermia causes CDK5 insolubilization and loss of tyrosine-15 phosphorylation, both of which were prevented in cells overexpressing HSP70. Inhibition of CDK5 activity with roscovitine-sensitized cells to heat induced apoptosis indicating a protective role for CDK5 in inhibiting heat-induced apoptosis. Both roscovitine and heat shock treatment caused increased accumulation of NOXA a pro-apoptotic BH3-only member of the BCL2 family. The increased abundance of NOXA by CDK5 inhibition was not a result of changes in NOXA protein turnover. Instead, CDK5 inhibition increased NOXA mRNA and protein levels by decreasing the expression of miR-23a, whereas overexpressing the CDK5 activator p35 attenuated both of these effects on NOXA and miR-23a expression. Lastly, overexpression of miR-23a prevented apoptosis under conditions in which CDK5 activity was inhibited. These results demonstrate that CDK5 activity provides resistance to heat-induced apoptosis through the expression of miR-23a and subsequent suppression of NOXA synthesis. Additionally, they indicate that hyperthermia induces apoptosis through the insolubilization and inhibition of CDK5 activity.
Collapse
Affiliation(s)
- Trevor M Morey
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Rabih Roufayel
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Donald S Johnston
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Andrew S Fletcher
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| | - Dick D Mosser
- From the Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario N1G 2W1, Canada
| |
Collapse
|
30
|
Ko KI, Coimbra LS, Tian C, Alblowi J, Kayal RA, Einhorn TA, Gerstenfeld LC, Pignolo RJ, Graves DT. Diabetes reduces mesenchymal stem cells in fracture healing through a TNFα-mediated mechanism. Diabetologia 2015; 58:633-642. [PMID: 25563724 PMCID: PMC4346353 DOI: 10.1007/s00125-014-3470-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 11/19/2014] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS Diabetes interferes with bone formation and impairs fracture healing, an important complication in humans and animal models. The aim of this study was to examine the impact of diabetes on mesenchymal stem cells (MSCs) during fracture repair. METHODS Fracture of the long bones was induced in a streptozotocin-induced type 1 diabetic mouse model with or without insulin or a specific TNFα inhibitor, pegsunercept. MSCs were detected with cluster designation-271 (also known as p75 neurotrophin receptor) or stem cell antigen-1 (Sca-1) antibodies in areas of new endochondral bone formation in the calluses. MSC apoptosis was measured by TUNEL assay and proliferation was measured by Ki67 antibody. In vitro apoptosis and proliferation were examined in C3H10T1/2 and human-bone-marrow-derived MSCs following transfection with FOXO1 small interfering (si)RNA. RESULTS Diabetes significantly increased TNFα levels and reduced MSC numbers in new bone area. MSC numbers were restored to normal levels with insulin or pegsunercept treatment. Inhibition of TNFα significantly reduced MSC loss by increasing MSC proliferation and decreasing MSC apoptosis in diabetic animals, but had no effect on MSCs in normoglycaemic animals. In vitro experiments established that TNFα alone was sufficient to induce apoptosis and inhibit proliferation of MSCs. Furthermore, silencing forkhead box protein O1 (FOXO1) prevented TNFα-induced MSC apoptosis and reduced proliferation by regulating apoptotic and cell cycle genes. CONCLUSIONS/INTERPRETATION Diabetes-enhanced TNFα significantly reduced MSC numbers in new bone areas during fracture healing. Mechanistically, diabetes-enhanced TNFα reduced MSC proliferation and increased MSC apoptosis. Reducing the activity of TNFα in vivo may help to preserve endogenous MSCs and maximise regenerative potential in diabetic patients.
Collapse
Affiliation(s)
- Kang I. Ko
- Department of Periodontics, University of Pennsylvania, 240 S 40th St, Levy 122 Philadelphia, PA19104, USA
| | - Leila S. Coimbra
- Department of Physiology and Pathology, Araraquara Dental School, State University of São Paulo, Araraquara, São Paulo , Brazil
| | - Chen Tian
- Department of Periodontics, University of Pennsylvania, 240 S 40th St, Levy 122 Philadelphia, PA19104, USA
| | - Jazia Alblowi
- Department of Oral Basic and Clinical Sciences, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rayyan A. Kayal
- Department of Oral Basic and Clinical Sciences, Faculty of Dentistry, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Thomas A. Einhorn
- Department of Orthopaedic Surgery, School of Medicine, Boston University, Boston, MA, USA
| | - Louis C. Gerstenfeld
- Department of Orthopaedic Surgery, School of Medicine, Boston University, Boston, MA, USA
| | - Robert J. Pignolo
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dana T. Graves
- Department of Periodontics, University of Pennsylvania, 240 S 40th St, Levy 122 Philadelphia, PA19104, USA
| |
Collapse
|
31
|
CXCL13 promotes the effect of bone marrow mesenchymal stem cells (MSCs) on tendon-bone healing in rats and in C3HIOT1/2 cells. Int J Mol Sci 2015; 16:3178-87. [PMID: 25647417 PMCID: PMC4346887 DOI: 10.3390/ijms16023178] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/14/2015] [Accepted: 01/21/2015] [Indexed: 01/08/2023] Open
Abstract
Objectives: Mesenchymal stem cells (MSCs) are potential effective therapy for tissue repair and bone regeneration. In present study, the effects of CXC chemokine ligand-13 (CXCL13) were evaluated on tendon-bone healing of rats. Methods: Tendon bone healing of the rat model was established and biomechanical testing was performed at 2, 4, 8 weeks after surgery. Murine mesenchymal cell line (C3HIOT1/2 cells) was cultured. The expression of miRNA-23a was detected by real-time PCR. The protein expression of ERK1/2, JNK and p38 was detected by western blotting. MiR-23a mimic and inhibitor were used to overexpress or silence the expression of miR-23a. Results: MSCs significantly elevated the levels of ultimate load to failure, stiffness and stress in specimens of rats, the effects of which were enhanced by CXCL13. The expression of miR-23a was down-regulated and the protein of ERK1/2 level was up-regulated by CXCL13 treatment in both in vivo and in vitro experiments. ERK1/2 expression was elevated by overexpression of miR-23a and reduced by miR-23a inhibitor. Conclusions: These findings revealed that CXCL13 promoted the tendon-bone healing in rats with MSCs treatment, and implied that the activation of ERK1/2 via miR-23a was involved in the process of MSCs treated bone regeneration.
Collapse
|
32
|
Roufayel R, Johnston DS, Mosser DD. The elimination of miR-23a in heat-stressed cells promotes NOXA-induced cell death and is prevented by HSP70. Cell Death Dis 2014; 5:e1546. [PMID: 25429623 PMCID: PMC4260742 DOI: 10.1038/cddis.2014.484] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Revised: 10/01/2014] [Accepted: 10/08/2014] [Indexed: 12/21/2022]
Abstract
Protein-damaging stress stimulates cell destruction through apoptosis; however, non-lethal proteotoxic stress induces an adaptive response leading to the increased synthesis of heat shock proteins, which inhibit apoptosis. In this study, we sought to determine the mechanism responsible for the accumulation of the BH3-only protein NOXA in heat-stressed cells and its prevention by the heat shock protein HSP70. Analysis of transcript levels by RT-qPCR revealed that miR-23a levels decreased in heat-stressed cells and that this was correlated with an increased abundance of NOXA mRNA, which contains a miR-23a binding site in its 3′ untranslated region. Cells overexpressing HSP70 had higher levels of miR-23a, maintained these levels after heat shock and accumulated lower levels of NOXA mRNA and protein. The enhanced abundance of mir-23a in these HSP70-expressing cells is primarily due to its increased stability although higher levels of pri/pre-miR-23a expression, nuclear export and maturation were also contributing factors. Stable overexpression of miR-23a in the acute lymphoblastic T-cell line PEER resulted in reduced basal and heat-induced levels of NOXA mRNA and significantly inhibited heat-induced apoptosis. Additionally, stable overexpression of an shRNA targeting miR-23a in U937 lymphoma cells produced stable knockdown of miR-23a and resulted in increased NOXA mRNA and an increased sensitivity to heat-induced apoptosis. These results demonstrate the novel finding that hyperthermia affects the abundance of a microRNA that targets the expression of a pro-apoptotic protein and that HSP70 protects cells from heat-induced apoptosis by regulating the abundance of this microRNA. We speculate that the inhibition of miRNA transcription in heat-stressed cells could represent a general mechanism for apoptosis induction that is regulated by the molecular chaperone protein HSP70. Furthermore, we propose that HSP70 could be beneficial to tumor cells by helping to maintain the expression of oncogenic miRNAs under conditions of cellular stress.
Collapse
Affiliation(s)
- R Roufayel
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - D S Johnston
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| | - D D Mosser
- Department of Molecular and Cellular Biology, University of Guelph, 50 Stone Road East, Guelph, Ontario, N1G 2W1, Canada
| |
Collapse
|
33
|
Xing Y, Hou J, Guo T, Zheng S, Zhou C, Huang H, Chen Y, Sun K, Zhong T, Wang J, Li H, Wang T. microRNA-378 promotes mesenchymal stem cell survival and vascularization under hypoxic-ischemic conditions in vitro. Stem Cell Res Ther 2014; 5:130. [PMID: 25418617 PMCID: PMC4446090 DOI: 10.1186/scrt520] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 11/12/2014] [Indexed: 12/18/2022] Open
Abstract
Introduction Mesenchymal stem cells (MSCs) transplantation has been demonstrated to be an effective strategy for the treatment of cardiovascular disease. However, the low survival rate of MSCs at local diseased tissue reduces the therapeutic efficacy. We therefore investigated the influence of MicroRNA-378 (miR-378) transfection on MSCs survival and vascularization under hypoxic-ischemic condition in vitro. Methods MSCs were isolated from bone marrow of Sprague–Dawley rats and cultured in vitro. The third passage of MSCs were divided into the miR-378 group and control group. For the miR-378 group, cells were transfected with miR-378 mimic. Both groups experienced exposure to hypoxia (1% O2) and serum deprivation for 24 hours, using normoxia (20% O2) as a negative control during the process. After 24 hours of reoxygenation (20% O2), cell proliferation and apoptosis were evaluated. Expressions of apoptosis and angiogenesis related genes were detected. Both groups were further co-cultured with human umbilical vein endothelial cells to promote vascular differentiation for another 6 hours. Vascular density was assessed thereafter. Results Compared with the control group, MSCs transfected with miR-378 showed more rapid growth. Their proliferation rates were much higher at 72 h and 96 h under hypoxic condition (257.33% versus 246.67%, P <0.01; 406.84% versus 365.39%, P <0.05). Cell apoptosis percentage in the miR-378 group was significantly declined under normoxic and hypoxic condition (0.30 ± 0.10% versus 0.50 ± 0.10%, P <0.05; 0.60 ± 0.40% versus 1.70 ± 0.20%, P <0.01). The miR-378 group formed a larger number of vascular branches on matrigel. BCL2 level was decreased accompanied with an upregulated expression of BAX in the two experimental groups under the hypoxic environment. BAX expression was reduced in the miR-378 group under the hypoxic environment. In the miR-378 group, there was a decreased expression of tumor necrosis factor-α on protein level and a reduction of TUSC-2 under normoxic environment. Their expressions were both downregulated under hypoxic environment. For the angiogenesis related genes, enhanced expressions of vascular endothelial growth factorα, platelet derived growth factor-β and transforming growth factor-β1 could be detected both in normoxic and hypoxic-ischemic conditions. Conclusion MiR-378 transfection could effectively promote MSCs survival and vascularization under hypoxic-ischemic condition in vitro.
Collapse
|
34
|
Zhao H, Tao Z, Wang R, Liu P, Yan F, Li J, Zhang C, Ji X, Luo Y. MicroRNA-23a-3p attenuates oxidative stress injury in a mouse model of focal cerebral ischemia-reperfusion. Brain Res 2014; 1592:65-72. [PMID: 25280466 DOI: 10.1016/j.brainres.2014.09.055] [Citation(s) in RCA: 92] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2014] [Revised: 09/12/2014] [Accepted: 09/23/2014] [Indexed: 11/24/2022]
Abstract
The present study was designed to investigate the potential role of miR-23a-3p in experimental brain ischemia-reperfusion injury. Cerebral ischemia reperfusion was induced by transient middle cerebral artery occlusion (MCAO) for 1h in C57/BL6 mice. And miR-23a-3p angomir was transfected to upregulate the miR-23a-3p level. Our results showed that miR-23a-3p levels were transiently increased at 4h after reperfusion in the peri-infarction area, while markedly increased in the infarction core at reperfusion 4h and 24h. Importantly, in vivo study demonstrated that miR-23a-3p angomir treatment through intracerebroventricular injection markedly decreased cerebral infarction volume after MCAO. Simultaneously, miR-23a-3p reduced peroxidative production nitric oxide (NO) and 3-nitrotyrosine (3-NT), and increased the expression of manganese superoxide dismutase (MnSOD). In vitro study demonstrated that miR-23a-3p decreased hydrogen peroxide (H2O2)-induced lactate dehydrogenase (LDH) leakage dose-dependently, and reduced protein levels of activated caspase-3 in neuro-2a cells. In addition, miR-23a-3p reduced H2O2-induced production of NO and 3-NT dose-dependently, and reversed the decreased activity of total SOD and MnSOD in neuro-2a cells. Our study indicated that miR-23a-3p suppressed oxidative stress and lessened cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Haiping Zhao
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Zhen Tao
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Rongliang Wang
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Ping Liu
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Feng Yan
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Jincheng Li
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China
| | - Chencheng Zhang
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 100053, China; Beijing Institute for Brain Disorders, Beijing 100053, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute, Xuanwu Hospital of Capital Medical University, 45 Changchun Street, Beijing 100053, China; Beijing Geriatric Medical Research Center, Beijing 100053, China; Beijing Institute for Brain Disorders, Beijing 100053, China; Key Laboratory of Neurodegenerative Diseases of Ministry of Education, Beijing 100053, China; Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 100053, China.
| |
Collapse
|
35
|
Boštjančič E, Glavač D. miRNome in myocardial infarction: Future directions and perspective. World J Cardiol 2014; 6:939-958. [PMID: 25276296 PMCID: PMC4176804 DOI: 10.4330/wjc.v6.i9.939] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 03/28/2014] [Accepted: 06/27/2014] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs), which are small and non-coding RNAs, are genome encoded from viruses to humans. They contribute to various developmental, physiological and pathological processes in living organisms. A huge amount of research results revealed that miRNAs regulate these processes also in the heart. miRNAs may have cell-type-specific or tissue-specific expression patterns or may be expressed ubiquitously. Primary studies of miRNA involvement in hypertrophy, heart failure and myocardial infarction analyzed miRNAs that are enriched in or specific for cardiomyocytes; however, growing evidence suggest that other miRNAs, not cardiac or muscle-specific, play a significant role in cardiovascular disease. Abnormal miRNA regulation has been shown to be involved in cardiac diseases, suggesting that miRNAs might affect cardiac structure and function. In this review, we focus on miRNAs that have been found to contribute to the pathogenesis of myocardial infarction (MI) and the response post-MI and characterized as diagnostic, prognostic and therapeutic targets. The majority of these studies were performed using mouse and rat models of MI, with a focus on the identification of basic cellular and molecular pathways involved in MI and in the response post-MI. Much research has also been performed on animal and human plasma samples from MI individuals to identify miRNAs that are possible prognostic and/or diagnostic targets of MI and other MI-related diseases. A large proportion of research is focused on miRNAs as promising therapeutic targets and biomarkers of drug responses and/or stem cell treatment approaches. However, only a few studies have described miRNA expression in human heart tissue following MI.
Collapse
|