1
|
Nevins S, McLoughlin CD, Oliveros A, Stein JB, Rashid MA, Hou Y, Jang MH, Lee KB. Nanotechnology Approaches for Prevention and Treatment of Chemotherapy-Induced Neurotoxicity, Neuropathy, and Cardiomyopathy in Breast and Ovarian Cancer Survivors. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300744. [PMID: 37058079 PMCID: PMC10576016 DOI: 10.1002/smll.202300744] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/05/2023] [Indexed: 06/19/2023]
Abstract
Nanotechnology has emerged as a promising approach for the targeted delivery of therapeutic agents while improving their efficacy and safety. As a result, nanomaterial development for the selective targeting of cancers, with the possibility of treating off-target, detrimental sequelae caused by chemotherapy, is an important area of research. Breast and ovarian cancer are among the most common cancer types in women, and chemotherapy is an essential treatment modality for these diseases. However, chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy are common side effects that can affect breast and ovarian cancer survivors quality of life. Therefore, there is an urgent need to develop effective prevention and treatment strategies for these adverse effects. Nanoparticles (NPs) have extreme potential for enhancing therapeutic efficacy but require continued research to elucidate beneficial interventions for women cancer survivors. In short, nanotechnology-based approaches have emerged as promising strategies for preventing and treating chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy. NP-based drug delivery systems and therapeutics have shown potential for reducing the side effects of chemotherapeutics while improving drug efficacy. In this article, the latest nanotechnology approaches and their potential for the prevention and treatment of chemotherapy-induced neurotoxicity, neuropathy, and cardiomyopathy in breast and ovarian cancer survivors are discussed.
Collapse
Affiliation(s)
- Sarah Nevins
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Callan D. McLoughlin
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Alfredo Oliveros
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Joshua B. Stein
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mohammad Abdur Rashid
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| | - Mi-Hyeon Jang
- Department of Neurosurgery, Robert Wood Johnson Medical
School, Rutgers University, the State University of New Jersey, 661 Hoes Ln W,
Piscataway, NJ, 08854, U.S.A
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers
University, the State University of New Jersey, 123 Bevier Road, Piscataway, NJ
08854, U.S.A
| |
Collapse
|
2
|
Zhao Q, Yang W, Li X, Yuan H, Guo J, Wang Y, Shan Z. MicroRNA-499-5p inhibits transforming growth factor-β1-induced Smad2 signaling pathway and suppresses fibroblast proliferation and collagen synthesis in rat by targeting TGFβ-R1. Mol Biol Rep 2023; 50:9757-9767. [PMID: 37676431 PMCID: PMC10676300 DOI: 10.1007/s11033-023-08755-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/10/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Artial fibrosis has been recognized as a typical pathological change in atrial fibrillation. Although present evidence suggests that microRNA-499-5p (miR-499-5p) plays an important role in the development of atrial fibrosis, the specific mechanism is not fully understood. Therefore, this study attempted to assess the influence of miR-499-5p on atrial fibroblasts and explore the potential molecular mechanism. METHODS Atrial fibroblasts from sprague dawley rat were respectively transfected with miR-499-5p mimic, miR-499-5p negative control and miR-499-5p inhibitor, atrial fibroblasts without any treatment were also established. Cell counting kit-8 assay and transwell assay were used to detect the proliferation and migration of atrial fibroblasts in each group. Expressions of miR-499-5p, TGF-β1, smad2, α-SMA, collagen-I and TGFβ-R1 in mRNA and protein level were subsequently detected via quantitative real-time polymerase chain reaction and western blot. Furthermore, the prediction of the binding sites of miR-499-5p and TGFβ-R1 was performed via the bioinformatics online software TargetScan and verified by dual luciferase reporter. RESULTS By utilizing miR-499-5p-transfected atrial fibroblasts model, expression of miR-499-5p in the miR-499-5p mimic group was upregulated, while it was downregulated in the miR-499-5p inhibitors group. Upregulated miR-499-5p expression led to to a significant decrease in the proliferative and migratory ability of cultured atrial fibroblasts, while downregulated miR-499-5p expression led to a significant increase in the proliferative and migratory ability of cultured atrial fibroblasts. Additionally, upregulated miR-499-5p expression made a significant rise in TGF-β1-induced mRNA and protein expression of TGF-β1, TGFβ-R1, smad2, α-SMA and collagen-I in atrial fibroblasts. Furthermore, results from the dual luciferase reporter conformed that miR-499-5p may repress TGFβ-R1 by binding the 3'UTR of TGFβ-R1 directly. CONCLUSIONS miR-499-5p is able to inhibit the activation of transforming growth factor β-induced Smad2 signaling and eventually suppressed the proliferation, migration and invasion of atrial fibroblasts and collagen synthesis by targeting TGFβ-R1.
Collapse
Affiliation(s)
- Qing Zhao
- Chinese PLA Medical Academy, Beijing, China
- Department of Cardiovascular Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Wentao Yang
- Department of Cardiology, Beijing Jishuitan Hospital, The Fourth Clinical Medical College of Peking University, Beijing, China
| | | | - Hongtao Yuan
- Department of Cardiovascular Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | | | - Yutang Wang
- Department of Geriatric Cardiology, Chinese PLA General Hospital, Beijing, China
| | - Zhaoliang Shan
- Chinese PLA Medical Academy, Beijing, China.
- Department of Cardiovascular Medicine, The Sixth Medical Center, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
3
|
Ravingerova T, Adameova A, Lonek L, Farkasova V, Ferko M, Andelova N, Kura B, Slezak J, Galatou E, Lazou A, Zohdi V, Dhalla NS. Is Intrinsic Cardioprotection a Laboratory Phenomenon or a Clinically Relevant Tool to Salvage the Failing Heart? Int J Mol Sci 2023; 24:16497. [PMID: 38003687 PMCID: PMC10671596 DOI: 10.3390/ijms242216497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 11/13/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Cardiovascular diseases, especially ischemic heart disease, as a leading cause of heart failure (HF) and mortality, will not reduce over the coming decades despite the progress in pharmacotherapy, interventional cardiology, and surgery. Although patients surviving acute myocardial infarction live longer, alteration of heart function will later lead to HF. Its rising incidence represents a danger, especially among the elderly, with data showing more unfavorable results among females than among males. Experiments revealed an infarct-sparing effect of ischemic "preconditioning" (IPC) as the most robust form of innate cardioprotection based on the heart's adaptation to moderate stress, increasing its resistance to severe insults. However, translation to clinical practice is limited by technical requirements and limited time. Novel forms of adaptive interventions, such as "remote" IPC, have already been applied in patients, albeit with different effectiveness. Cardiac ischemic tolerance can also be increased by other noninvasive approaches, such as adaptation to hypoxia- or exercise-induced preconditioning. Although their molecular mechanisms are not yet fully understood, some noninvasive modalities appear to be promising novel strategies for fighting HF through targeting its numerous mechanisms. In this review, we will discuss the molecular mechanisms of heart injury and repair, as well as interventions that have potential to be used in the treatment of patients.
Collapse
Affiliation(s)
- Tanya Ravingerova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Adriana Adameova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University in Bratislava, 10 Odbojárov St., 832 32 Bratislava, Slovakia
| | - Lubomir Lonek
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Veronika Farkasova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Miroslav Ferko
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Natalia Andelova
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Branislav Kura
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Jan Slezak
- Institute for Heart Research, Centre of Experimental Medicine, Slovak Academy of Sciences, 9 Dubravska cesta, 841 04 Bratislava, Slovakia; (A.A.); (L.L.); (V.F.); (M.F.); (N.A.); (B.K.); (J.S.)
| | - Eleftheria Galatou
- School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (E.G.); (A.L.)
- Department of Life and Health Sciences, University of Nicosia, 2417 Nicosia, Cyprus
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, 541 24 Thessaloniki, Greece; (E.G.); (A.L.)
| | - Vladislava Zohdi
- Department of Anatomy, Faculty of Medicine, Comenius University in Bratislava, 24 Špitalska, 813 72 Bratislava, Slovakia;
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, 19 Innovation Walk, Clayton, VIC 3800, Australia
| | - Naranjan S. Dhalla
- Institute of Cardiovascular Sciences St. Boniface Hospital Albrechtsen Research Centre, 351 Tache Avenue, Winnipeg, MB R2H 2A6, Canada;
| |
Collapse
|
4
|
Al-Masri A. Apoptosis and long non-coding RNAs: Focus on their roles in Heart diseases. Pathol Res Pract 2023; 251:154889. [PMID: 38238070 DOI: 10.1016/j.prp.2023.154889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 01/23/2024]
Abstract
Heart disease is one of the principal death reasons around the world and there is a growing requirement to discover novel healing targets that have the potential to avert or manage these illnesses. On the other hand, apoptosis is a strongly controlled, cell removal procedure that has a crucial part in numerous cardiac problems, such as reperfusion injury, MI (myocardial infarction), consecutive heart failure, and inflammation of myocardium. Completely comprehending the managing procedures of cell death signaling is critical as it is the primary factor that influences patient mortality and morbidity, owing to cardiomyocyte damage. Indeed, the prevention of heart cell death appears to be a viable treatment approach for heart illnesses. According to current researches, a number of long non-coding RNAs cause the heart cells death via different methods that are embroiled in controlling the activity of transcription elements, the pathways that signals transmission within cells, small miRNAs, and the constancy of proteins. When there is too much cell death in the heart, it can cause problems like reduced blood flow, heart damage after restoring blood flow, heart disease in diabetics, and changes in the heart after reduced blood flow. Therefore, studying how lncRNAs control apoptosis could help us find new treatments for heart diseases. In this review, we present recent discoveries about how lncRNAs are involved in causing cell death in different cardiovascular diseases.
Collapse
Affiliation(s)
- Abeer Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
5
|
Jin Y, Zhang Q, Liu Y, Guan H, Shou Y, Du C, Luo J, Zhao K, Li C, Xu L, Ma W, Chen N, Zheng Y, Yu D. Identifying microRNAs that drive BaP-induced pulmonary effects: Multiple patterns of mechanisms underlying activation of the toxicity pathways. ENVIRONMENT INTERNATIONAL 2022; 170:107588. [PMID: 36274491 DOI: 10.1016/j.envint.2022.107588] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/14/2022] [Accepted: 10/14/2022] [Indexed: 06/16/2023]
Abstract
MiRNAs are widely acknowledged as regulating gene expression and thus, being involved in broad biological functions, environmental responses, and the process of diseases. Epidemiology could provide exposure- or disease-relevant miRNAs, while toxicology could reveal the underlying mechanisms. Here, a new "Bottom-up" approach was proposed to identify miRNAs that are responsible for environmental exposure-induced adverse outcomes. In our previous study, 5 key toxicity pathways were established underlying BaP-induced lung diseases; further, genes from these 5 pathways that were responsive to BaP exposure in HBE-CYP1A1 cells were identified. In this study, we identified 26 miRNA:mRNA interactions during BaP exposure through RNA-sequencing using the same HBE-CYP1A1 cells. According to the expression alteration and regulatory mechanisms, we summarized 8 action patterns of miRNA:mRNA, which led to the induction of miRNAs that predominantly regulate target genes and responsible are for the pathway perturbations (as "drivers"), and miRNAs that subordinately regulate genes during pathway perturbations (as "symptoms"). 5 corresponding miRNAs: miR-3173-5p, miR-629-3p, miR-9-5p, miR-1343-3p and miR-219a-1-3p were identified as "drivers", and were all validated with expression alteration in lung disease patients from published studies. In conclusion, this study offers a new approach to identification of epigenetic factors that may shed light on the causation of environment-related health outcomes.
Collapse
Affiliation(s)
- Yuan Jin
- School of Public Health, Qingdao University, Qingdao, China
| | - Qianqian Zhang
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuzhen Liu
- School of Public Health, Qingdao University, Qingdao, China
| | - Heyuan Guan
- School of Public Health, Qingdao University, Qingdao, China
| | - Yingqing Shou
- School of Public Health, Qingdao University, Qingdao, China
| | - Chenlong Du
- School of Public Health, Qingdao University, Qingdao, China
| | - Jiao Luo
- School of Public Health, Qingdao University, Qingdao, China
| | - Kunming Zhao
- School of Public Health, Qingdao University, Qingdao, China
| | - Chuanhai Li
- School of Public Health, Qingdao University, Qingdao, China
| | - Lin Xu
- School of Public Health, Qingdao University, Qingdao, China
| | - Wanli Ma
- School of Public Health, Qingdao University, Qingdao, China
| | - Ningning Chen
- School of Public Health, Qingdao University, Qingdao, China
| | - Yuxin Zheng
- School of Public Health, Qingdao University, Qingdao, China
| | - Dianke Yu
- School of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
6
|
Ghosh N, Fenton S, van Hout I, Jones GT, Coffey S, Williams MJA, Sugunesegran R, Parry D, Davis P, Schwenke DO, Chatterjee A, Katare R. Therapeutic knockdown of miR-320 improves deteriorated cardiac function in a pre-clinical model of non-ischemic diabetic heart disease. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 29:330-342. [PMID: 35950211 PMCID: PMC9356207 DOI: 10.1016/j.omtn.2022.07.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/08/2022] [Indexed: 11/17/2022]
Abstract
Non-ischemic diabetic heart disease (NiDHD) is characterized by diastolic dysfunction and decreased or preserved systolic function, eventually resulting in heart failure. Accelerated apoptotic cell death because of alteration of molecular signaling pathways due to dysregulation in microRNAs (miRNAs) plays a significant role in the development of NiDHD. Here, we aimed to determine the pathological role of cardiomyocyte-enriched pro-apoptotic miR-320 in the development of NiDHD. We identified a marked upregulation of miR-320 that was associated with downregulation of its target protein insulin growth factor-1 (IGF-1) in human right atrial appendage tissue in the late stages of cardiomyopathy in type 2 diabetic db/db mice and high-glucose-cultured human ventricular cardiomyocytes (AC-16 cells). In vitro knockdown of miR-320 in high-glucose-exposed AC-16 cells using locked nucleic acid (LNA) anti-miR-320 markedly reduced high-glucose-induced apoptosis by restoring IGF-1 and Bcl-2. Finally, in vivo knockdown of miR-320 in 24-week-old type 2 diabetic db/db mice reduced cardiomyocyte apoptosis and interstitial fibrosis while restoring vascular density. This resulted in partial recovery of the impaired diastolic and systolic function. Our study provides evidence that miR-320 is a late-responding miRNA that aggravates apoptosis and cardiac dysfunction in the diabetic heart, and that therapeutic knockdown of miR-320 is beneficial in partially restoring the deteriorated cardiac function.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Sonya Fenton
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Isabelle van Hout
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Gregory T Jones
- Department of Surgical Sciences, University of Otago, Dunedin, New Zealand
| | - Sean Coffey
- Department of Medicine, University of Otago, Dunedin, New Zealand
| | | | | | - Dominic Parry
- Department of Cardiothoracic Surgery, University of Otago, Dunedin, New Zealand
| | - Philip Davis
- Department of Cardiothoracic Surgery, University of Otago, Dunedin, New Zealand
| | - Daryl O Schwenke
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Anirudha Chatterjee
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand.,Honorary Professor, UPES University, Dehradun, India
| | - Rajesh Katare
- Department of Physiology-HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
7
|
MiRNA-122-5p inhibitor abolishes angiotensin II-mediated loss of autophagy and promotion of apoptosis in rat cardiofibroblasts by modulation of the apelin-AMPK-mTOR signaling. In Vitro Cell Dev Biol Anim 2022; 58:136-148. [PMID: 35133561 DOI: 10.1007/s11626-022-00651-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2021] [Indexed: 11/05/2022]
Abstract
MicroRNAs (miRNAs) have emerged as essential regulators that could have pivotal roles in cardiac homeostasis and pathological remodeling of various cardiovascular diseases. We previously demonstrated that miRNA-122-5p overexpression exacerbated the process of vascular hypertrophy, fibrosis, and dysfunction in hypertensive rats and rat aortic adventitial fibroblasts. However, the exact roles and underlying mechanisms of miRNA-122-5p in myocardial fibroblasts remain largely unknown. In this work, neonatal rat cardiofibroblasts (CFs) were isolated and primarily cultured from the hearts of 2- to 3-d-old Sprague-Dawley rats. Stimulation of angiotensin II (Ang II) resulted in marked increases in cellular proliferation and migration and levels of collagen I, collagen III, CTGF, and TGF-β1 in cultured CFs. Furthermore, Ang II led to promoted expression of P62, Bax, and phosphorylated mTOR as well as downregulation of LC3II, beclin-1, and AMPK-phosphorylated levels, thereby contributing to imbalance of autophagy and apoptosis, and cellular injury in CFs, which were significantly ameliorated by treatment with miRNA-122-5p inhibitor. These changes were associated with decreased levels of collagen I, collagen III, CTGF, and TGF-β1. Furthermore, Ang II-induced loss of autophagy and promotion of apoptosis in CFs were prevented by the treatment with Pyr1-apelin-13 or AMPK agonist AICAR or mTOR inhibitor rapamycin, respectively. In contrast, administration of miRNA-122-5p mimics and autophagy inhibitor 3-methylademine reversed beneficial roles of Pyr1-apelin-13. Collectively, these data indicated that miRNA-122-5p is an essential regulator of autophagy and apoptosis in rat CFs via the apelin/AMPK/mTOR signaling pathway, which may be potentially used as a therapeutic target in myocardial fibrosis and related diseases.
Collapse
|
8
|
Abstract
Chronic kidney disease (CKD), which is characterized by the gradual loss of kidney function, is a growing worldwide problem due to CKD-related morbidity and mortality. There are no reliable and early biomarkers enabling the monitoring, the stratification of CKD progression and the estimation of the risk of CKD-related complications, and therefore, the search for such molecules is still going on. Numerous studies have provided evidence that miRNAs are potentially important particles in the CKD field. Studies indicate that some miRNA levels can be increased in patients with CKD stages III–V and hemodialysis and decreased in renal transplant recipients (miR-143, miR-145 and miR-223) as well as elevated in patients with CKD stages III–V, decreased in hemodialysis patients and even more markedly decreased in renal transplant recipients (miR-126 and miR-155). miRNA have great potential of being sensitive and specific biomarkers in kidney diseases as they are tissue specific and stable in various biological materials. Some promising non-invasive miRNA biomarkers have already been recognized in renal disease with the potential to enhance diagnostic accuracy, predict prognosis and monitor the course of disease. However, large-scale clinical trials enrolling heterogeneous patients are required to evaluate the clinical value of miRNAs.
Collapse
|
9
|
Wang X, Yuan T, Yin N, Ma X, Yang Y, Yang J, Shaukat A, Deng G. Interferon-τ regulates the expression and function of bovine leukocyte antigen by downregulating bta-miR-204. Exp Ther Med 2021; 21:594. [PMID: 33884032 PMCID: PMC8056107 DOI: 10.3892/etm.2021.10026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 10/15/2020] [Indexed: 12/21/2022] Open
Abstract
IFN-τ is a pregnancy recognition factor that regulates embryo implantation in ruminants. IFN-τ has been suggested to be involved in the expression of microRNA (miRNA/miR) and bovine leukocyte antigen (BoLA), which is an analog of the human major histocompatibility complex class I. However, little is known about whether the miRNAs are involved in the expression of BoLA in ruminants. The present study firstly verified that bta-miR-204 was downregulated and that BoLA was upregulated in the uterine tissues of dairy cows during early pregnancy. Subsequently, luciferase reporter assays, reverse transcription-quantitative PCR and western blot analysis were used to validate BoLA as the target gene of bta-miR-204. Moreover, BoLA was markedly upregulated and bta-miR-204 was downregulated in bovine endometrial epithelial cells (bEECs) treated with IFN-τ. In addition, the results indicated that when the expression level of BoLA was increased by IFN-τ, the expression level of programmed death-ligand 1 (PD-L1) and programmed death-ligand 2 (PD-L2) was also increased. Furthermore, when BoLA was silenced in bEECs by small interfering RNA, the expression of PD-L1 and PD-L2 was not affected by IFN-τ. The expression level of PD-L1 and PD-L2 was also increased in the uterine tissues of pregnant dairy cattle. In conclusion, IFN-τ may function by suppressing the expression of bta-miR-204 to increase the expression of BoLA during the embryo implantation period in cattle. IFN-τ may induce PD-L1 and PD-L2 transcription by regulating BoLA, which may influence the T cell immune response, thereby regulating pregnant cattle immunization.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China.,College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing 404155, P.R. China
| | - Ting Yuan
- College of Animal Science and Technology, Chongqing Three Gorges Vocational College, Chongqing 404155, P.R. China
| | - Nannan Yin
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Xiaofei Ma
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Yaping Yang
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Jing Yang
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Aftab Shaukat
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| | - Ganzhen Deng
- Department of Clinical Veterinary Medicine, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei 430070, P.R. China
| |
Collapse
|
10
|
Xiang Q, Wang J, Wang T, Zuo H. Combination of baicalein and miR-106a-5p mimics significantly alleviates IL-1β-induced inflammatory injury in CHON-001 cells. Exp Ther Med 2021; 21:345. [PMID: 33732318 PMCID: PMC7903477 DOI: 10.3892/etm.2021.9776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) induces inflammation and degeneration of all joint components, and as such, is a considerable source of disability, pain and socioeconomic burden worldwide. Baicalein (BAI) and microRNA (miR)-106a-5p suppress the progression of OA; however, the effects of BAI and miR-106a-5p for the combined treatment of OA are not completely understood. An in vitro OA model was established by treating CHON-001 cells with 20 ng/ml interleukin (IL)-1β. Cell Counting Kit-8 and flow cytometry assays were conducted to evaluate cell viability and apoptosis, respectively. Western blotting was performed to determine the expression levels of Bax, active caspase-3, Bcl-2, collagen I, collagen III, aggrecan, matrix metallopeptidase (MMP)-13, MMP-9, active Notch1 and transcription factor hes family bHLH transcription factor 1 (Hes1). The levels of IL-6 and tumor necrosis factor-α in the cell culture medium were quantified via ELISA. The present study revealed that treatment with BAI or miR-106a-5p mimic alleviated IL-1β-induced apoptosis, and BAI + miR-106a-5p combination treatment exerted enhanced anti-inflammatory effects compared with monotherapy. Furthermore, IL-1β-induced accumulation of collagen, collagen III, MMP-13 and MMP-9 in CHON-001 cells was reversed to a greater degree following combination treatment compared with monotherapy. Likewise, IL-1β-induced aggrecan degradation was markedly reversed by combination treatment. IL-1β-induced upregulation of active Notch1 and Hes1 in CHON-001 cells was also significantly attenuated by combined BAI + miR-106a-5p treatment. In conclusion, the results of the present study revealed that the combination of BAI and miR-106a-5p mimic significantly decreased IL-1β-induced inflammatory injury in CHON-001 cells, which may serve as a novel therapeutic strategy for OA.
Collapse
Affiliation(s)
- Qingtian Xiang
- Department of Orthopedic Surgery, Xiang Shui County People's Hospital, Yancheng, Jiangsu 224600, P.R. China
| | - Jijun Wang
- Department of Orthopedic Surgery, Xiang Shui County People's Hospital, Yancheng, Jiangsu 224600, P.R. China
| | - Tongwei Wang
- Department of Orthopedic Surgery, Xiang Shui County People's Hospital, Yancheng, Jiangsu 224600, P.R. China
| | - Hongguang Zuo
- Department of Orthopedic Surgery, Xiang Shui County People's Hospital, Yancheng, Jiangsu 224600, P.R. China
| |
Collapse
|
11
|
Bian Z, Huang X, Chen Y, Meng J, Feng X, Zhang M, Zhang L, Zhou J, Liang C. Fifteen-MiRNA-Based Signature Is a Reliable Prognosis-Predicting Tool for Prostate Cancer Patients. Int J Med Sci 2021; 18:284-294. [PMID: 33390797 PMCID: PMC7738977 DOI: 10.7150/ijms.49412] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/02/2020] [Indexed: 12/09/2022] Open
Abstract
Recurrence is a major problem for prostate cancer patients, thus, identifying prognosis-related markers to evaluate clinical outcomes is essential. Here, we established a fifteen-miRNA-based recurrence-free survival (RFS) predicting signature based on the miRNA expression profile extracted from The Cancer Genome Atlas (TCGA) database by the LASSO Cox regression analysis. The median risk score generated by the signature in both the TCGA training and the external Memorial Sloan-Kettering Cancer Center (MSKCC) validation cohorts was employed and the patients were subclassified into low- and high-risk subgroups. The Kaplan-Meier plot and log-rank analyses showed significant survival differences between low- and high-risk subgroups of patients (TCGA, log-rank P < 0.001 & MSKCC, log-rank P = 0.045). In addition, the receiver operating characteristic curves of both the training and external validation cohorts indicated the good performance of our model. After predicting the downstream genes of these miRNAs, the miRNA-mRNA network was visualized by Cytoscape software. In addition, pathway analyses found that the differences between two groups were mainly enriched on tumor progression and drug resistance-related pathways. Multivariate analyses revealed that the miRNA signature is an independent indicator of RFS prognosis for prostate cancer patients with or without clinicopathological features. In summary, our novel fifteen-miRNA-based prediction signature is a reliable method to evaluate the prognosis of prostate cancer patients.
Collapse
Affiliation(s)
- Zichen Bian
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China
| | - Xinbo Huang
- Guangdong and Shenzhen Key Laboratory of Male Reproductive Medicine and Genetics, Institute of Urology, Peking University Shenzhen Hospital, Shenzhen-Peking University-the Hong Kong University of Science and Technology Medical Center, Shenzhen 518000, China
| | - Yiding Chen
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China
| | - Jialin Meng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China
| | - Xingliang Feng
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China
| | - Meng Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China.,Institute of Urology of Shenzhen University, The Third Affiliated Hospital of Shenzhen University, Shenzhen Luohu Hospital Group, Shenzhen 518000, People's Republic of China
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University and Institute of Urology and Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University, Jixi Road 218th, Shushan District, Hefei, Anhui, 230022, People's Republic of China
| |
Collapse
|
12
|
Li Z, Yi N, Chen R, Meng Y, Wang Y, Liu H, Cao W, Hu Y, Gu Y, Tong C, Lu M, Li L, Peng L. miR-29b-3p protects cardiomyocytes against endotoxin-induced apoptosis and inflammatory response through targeting FOXO3A. Cell Signal 2020; 74:109716. [PMID: 32707074 DOI: 10.1016/j.cellsig.2020.109716] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 07/12/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Cardiac dysfunction represents a main component of death induced by sepsis in critical care units. And microRNAs (miRNAs) have been reported as important modulators or biomarkers of sepsis. However, the molecular detail of miRNAs involved in septic cardiac dysfunction remains unclear. Here we showed that endotoxin (lipopolysaccharide, LPS) significantly down-regulated expression of miR-29b-3p in heart. Increased expression of miR-29b-3p by lentivirus improved cardiac function and attenuated damage of cardiac induced by LPS in mice. Furthermore, overexpression or knockdown of miR-29b-3p showed its crucial roles on regulation of apoptosis and production of pro-inflammatory cytokines in NRCMs through directly targeting FOXO3A. miR-29b-3p ameliorates inflammatory damage likely via reducing activation of MAPKs and nuclear-translocation of NF-κB to block LPS-activated NF-κB signaling. Notably, miR-29b is also down-regulated in septic patients' plasma compared with normal subjects, indicating a potential clinical relevance of miR-29b. Taken together, our findings demonstrate that upregulation of miR-29b-3p can attenuate myocardial injury induced by sepsis via regulating FOXO3A, which provide a potential therapy target for interference of septic cardiac dysfunction.
Collapse
Affiliation(s)
- Zhigang Li
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China
| | - Na Yi
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China
| | - Rou Chen
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Yilei Meng
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China
| | - Yumei Wang
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Huan Liu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China
| | - Wenze Cao
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China
| | - Yi Hu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China
| | - Yanqiong Gu
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Chang Tong
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Min Lu
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China
| | - Li Li
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai 200092, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China.
| | - Luying Peng
- Key Laboratory of Arrhythmias, Ministry of Education, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Institute of Medical Genetics, Tongji University, Shanghai 200092, China; Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Heart Health Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, China; Research Units of Origin and Regulation of Heart Rhythm, Chinese Academy of Medical Sciences, Shanghai 200092, China; Department of Medical Genetics, Tongji University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
13
|
An Overview of Non-coding RNAs and Cardiovascular System. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1229:3-45. [PMID: 32285403 DOI: 10.1007/978-981-15-1671-9_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cardiovascular disease management and timely diagnosis remain a major dilemma. Delineating molecular mechanisms of cardiovascular diseases is opening horizon in the field of molecular medicines and in the development of early diagnostic markers. Non-coding RNAs are the highly functional and vibrant nucleic acids and are known to be involved in the regulation of endothelial cells, vascular and smooth muscles cells, cardiac metabolism, ischemia, inflammation and many processes in cardiovascular system. This chapter is comprehensively focusing on the overview of the non-coding RNAs including their discovery, generation, classification and functional regulation. In addition, overview regarding different non-coding RNAs as long non-coding, siRNAs and miRNAs involvement in the cardiovascular diseases is also addressed. Detailed functional analysis of this vast group of highly regulatory molecules will be promising for shaping future drug discoveries.
Collapse
|
14
|
Wang W, Liu Q, Zhang T, Chen L, Li S, Xu S. Glyphosate induces lymphocyte cell dysfunction and apoptosis via regulation of miR-203 targeting of PIK3R1 in common carp (Cyprinus carpio L.). FISH & SHELLFISH IMMUNOLOGY 2020; 101:51-57. [PMID: 32217141 DOI: 10.1016/j.fsi.2020.03.047] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/12/2020] [Accepted: 03/21/2020] [Indexed: 06/10/2023]
Abstract
Glyphosate is a widely used pesticide worldwide. The problem surrounding glyphosate is worth investigating, especially with its increased use, and an increasing number of studies have found that the toxic effect of glyphosate is objective. MiR-203 was seldom found in fish diseases or glyphosate researches. This article aims to explore the effect of miR-203 on carp lymphocytes during glyphosate exposure. Therefore, acridine orange/ethidium bromide (AO/EB) and flow cytometry were carried out to evaluate apoptosis, and we also detected CYPs (CYP1A1, CYP1B1, CYP1C), cytokine secretion (IL-1β, IL-8, IL-10, IFN-γ, TNF-α), inflammatory factors (NF-κB, cox-2), and the expression of miR-203 and the PI3K/AKT pathway by RT-PCR and Western blot analyses. Our results demonstrated that glyphosate exposure could induce lymphocyte apoptosis via regulation of miR-203 targeting of PI3K/AKT, which was accompanied by CYPs activation, abnormal cytokine expression and an inflammatory response. These results show that glyphosate is not nontoxic to fish and provide new insights for the usage of glyphosate as an herbicide in the future.
Collapse
Affiliation(s)
- Wei Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Qingqing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Tianyi Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Lu Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China
| | - Shu Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China; Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, 150030, PR China.
| |
Collapse
|
15
|
Zhang H, Huang X, Liu J, Liu B. Simultaneous and ultrasensitive detection of multiple microRNAs by single-molecule fluorescence imaging. Chem Sci 2020; 11:3812-3819. [PMID: 34122849 PMCID: PMC8152581 DOI: 10.1039/d0sc00580k] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 03/23/2020] [Indexed: 11/24/2022] Open
Abstract
Cell status changes are typically accompanied by the simultaneous changes of multiple microRNA (miRNA) levels. Thus, simultaneous and ultrasensitive detection of multiple miRNA biomarkers shows great promise in early cancer diagnosis. Herein, a facile single-molecule fluorescence imaging assay was proposed for the simultaneous and ultrasensitive detection of multiple miRNAs using only one capture anti-DNA/RNA antibody (S9.6 antibody). Two complementary DNAs (cDNAs) designed to hybridize with miRNA-21 and miRNA-122 were labelled with Cy3 (cDNA1) and Cy5 (cDNA2) dyes at their 5'-ends, respectively. After hybridization, both miRNA-21/cDNA1 and miRNA-122/cDNA2 complexes were captured by S9.6 antibodies pre-modified on a coverslip surface. Subsequently, the Cy3 and Cy5 dyes on the coverslip surface were imaged by the single-molecule fluorescence setup. The amount of miRNA-21 and miRNA-122 was quantified by counting the image spots from the Cy3 and Cy5 dye molecules in the green and red channels, respectively. The proposed assay displayed high specificity and sensitivity for singlet miRNA detection both with a detection limit of 5 fM and for multiple miRNA detection both with a detection limit of 20 fM. Moreover, it was also demonstrated that the assay could be used to detect multiple miRNAs simultaneously in human hepatocellular cancer cells (HepG2 cells). The proposed assay provides a novel biosensing platform for the ultrasensitive and simple detection of multiple miRNA expressions and shows great prospects for early cancer diagnosis.
Collapse
Affiliation(s)
- Hongding Zhang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Institute of Biomedical Sciences, Fudan University Shanghai 200438 P. R. China
- College of Chemistry and Chemical Engineering, Xinyang Normal University Xinyang 464000 P. R. China
| | - Xuedong Huang
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Institute of Biomedical Sciences, Fudan University Shanghai 200438 P. R. China
| | - Jianwei Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Institute of Biomedical Sciences, Fudan University Shanghai 200438 P. R. China
| | - Baohong Liu
- Department of Chemistry, Shanghai Stomatological Hospital, State Key Laboratory of Molecular Engineering of Polymers, Institute of Biomedical Sciences, Fudan University Shanghai 200438 P. R. China
| |
Collapse
|
16
|
Felekkis K, Papaneophytou C. Challenges in Using Circulating Micro-RNAs as Biomarkers for Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21020561. [PMID: 31952319 PMCID: PMC7013987 DOI: 10.3390/ijms21020561] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
Micro-RNAs (miRNAs) play a pivotal role in the development and physiology of the cardiovascular system while they have been associated with multiple cardiovascular diseases (CVDs). Several cardiac miRNAs are detectable in circulation (circulating miRNAs; c-miRNAs) and are emerging as diagnostic and therapeutic biomarkers for CVDs. c-miRNAs exhibit numerous essential characteristics of biomarkers while they are extremely stable in circulation, their expression is tissue-/disease-specific, and they can be easily detected using sequence-specific amplification methods. These features of c-miRNAs are helpful in the development of non-invasive assays to monitor the progress of CVDs. Despite significant progress in the detection of c-miRNAs in serum and plasma, there are many contradictory publications on the alterations of cardiac c-miRNAs concentration in circulation. The aim of this review is to examine the pre-analytical and analytical factors affecting the quantification of c-miRNAs and provide general guidelines to increase the accuracy of the diagnostic tests in order to improve future research on cardiac c-miRNAs.
Collapse
|
17
|
Xiong W, Qu Y, Chen H, Qian J. Insight into long noncoding RNA-miRNA-mRNA axes in myocardial ischemia-reperfusion injury: the implications for mechanism and therapy. Epigenomics 2019; 11:1733-1748. [PMID: 31701757 DOI: 10.2217/epi-2019-0119] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Emerging evidence has demonstrated that regulatory noncoding RNAs (ncRNAs), such as long noncoding RNAs (lncRNAs) and miRNAs, play crucial roles in the initiation and progress of myocardial ischemia-reperfusion injury (MIRI), which is associated with autophagy, apoptosis and necrosis of cardiomyocytes, as well as oxidative stress, inflammation and mitochondrial dysfunction. LncRNAs serve as a precursor or host of miRNAs and directly/indirectly affecting miRNAs via competitive binding or sponge effects. Simultaneously, miRNAs post-transcriptionally regulate the expression of genes by targeting various mRNA sequences due to their imperfect pairing with mRNAs. This review summarizes the potential regulatory role of lncRNA-miRNA-mRNA axes in MIRI and related molecular mechanisms of cardiac disorders, also provides insight into the potential therapies for MIRI-induced diseases.
Collapse
Affiliation(s)
- Wei Xiong
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China
| | - Yan Qu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China.,Department of Anesthesiology, The Fourth Affiliated Hospital of Kunming Medical University, The Second People's Hospital of Yunnan, Kunming, Yunnan province 650021, PR China
| | - Hongmei Chen
- Department of Anesthesiology, Kunming Angel Women's & Children's Hospital, Kunming, Yunnan province 650108, PR China
| | - Jinqiao Qian
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan province 650032, PR China
| |
Collapse
|
18
|
Wu K, Zhao Q, Li Z, Li N, Xiao Q, Li X, Zhao Q. Bioinformatic screening for key miRNAs and genes associated with myocardial infarction. FEBS Open Bio 2018; 8:897-913. [PMID: 29928570 PMCID: PMC5985982 DOI: 10.1002/2211-5463.12423] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/07/2018] [Accepted: 03/23/2018] [Indexed: 12/11/2022] Open
Abstract
Despite significant advances in understanding of the causes of and treatment of myocardial infarction (MI) in recent years, morbidity and mortality is still high. The aim of this study was to identify miRNA and genes potentially associated with MI. mRNA and miRNA expression datasets were downloaded from the Gene Expression Omnibus database (http://www.ncbi.nlm.nih.gov/geo/). Interactions between miRNA and the expression and function of target genes were analyzed, and a protein–protein interaction network was constructed. The diagnostic value of identified miRNA and genes was assessed. Quantitative RT‐PCR was applied to validate the results of the bioinformatics analysis. MiR‐27a, miR‐31*, miR‐1291, miR‐139‐5p, miR‐204, miR‐375, and target genes including CX3CR1,HSPA6, and TPM3 had potential diagnostic value. The genes TFEB,IRS2,GRB2,FASLG,LIMS1,CX3CR1,HSPA6,TPM3,LAT2,CEBPD,AQP9, and MAPKAPK2 were associated with recovery from MI. In conclusion, the identified miRNA and genes might be associated with the pathology of MI.
Collapse
Affiliation(s)
- Ke Wu
- Department of Cardiology Beijing Anzhen Hospital Capital Medical University Beijing China.,Department of Cardiology Central Hospital of Taian of Shandong Province China
| | - Qiang Zhao
- Department of Cardiology Affiliated Hospital of Taishan Medical University of Shandong Province Taian China
| | - Zhengmei Li
- Department of Radiology Taishan Medical University of Shandong Province Taian China
| | - Nannan Li
- Department of Respiration Medicine Central Hospital of Taian of Shandong Province China
| | - Qiang Xiao
- Department of Cardiology Affiliated Hospital of Taishan Medical University of Shandong Province Taian China
| | - Xiuchang Li
- Department of Cardiology Affiliated Hospital of Taishan Medical University of Shandong Province Taian China
| | - Quanming Zhao
- Department of Cardiology Beijing Anzhen Hospital Capital Medical University Beijing China
| |
Collapse
|
19
|
Ultimo S, Zauli G, Martelli AM, Vitale M, McCubrey JA, Capitani S, Neri LM. Cardiovascular disease-related miRNAs expression: potential role as biomarkers and effects of training exercise. Oncotarget 2018; 9:17238-17254. [PMID: 29682219 PMCID: PMC5908320 DOI: 10.18632/oncotarget.24428] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 01/20/2018] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are one of the most important causes of mortality worldwide, therefore the need of effective preventive strategies is imperative. Aging is associated with significant changes in both cardiovascular structure and function that lower the threshold for clinical signs and symptoms, making older people more susceptible to CVDs morbidity and mortality. microRNAs (miRNAs) modulate gene expression at post-transcriptional level and increasing evidence has shown that miRNAs are involved in cardiovascular physiology and in the pathogenesis of CVDs. Physical activity is recommended by the medical community and the cardiovascular benefits of exercise are multifactorial and include important systemic effects on skeletal muscle, the peripheral vasculature, metabolism, and neuroendocrine systems, as well as beneficial modifications within the myocardium itself. In this review we describe the role of miRNAs and their dysregulation in several types of CVDs. We provide an overview of miRNAs in CVDs and of the effects of physical activity on miRNA regulation involved in both cardiovascular pathologies and age-related cardiovascular changes and diseases. Circulating miRNAs in response to acute and chronic sport exercise appear to be modulated following training exercise, and may furthermore serve as potential biomarkers for CVDs and different age-related CVDs.
Collapse
Affiliation(s)
- Simona Ultimo
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Giorgio Zauli
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Vitale
- Department of Medicine and Surgery, University of Parma, Parma, Italy.,CoreLab, Azienda Ospedaliero, Universitaria di Parma, Parma, Italy
| | - James A McCubrey
- Department of Microbiology and Immunology, Brody School of Medicine, East Carolina University, Greenville, NC, USA
| | - Silvano Capitani
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Luca M Neri
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| |
Collapse
|
20
|
Ji Q, Qi D, Xu X, Xu Y, Goodman SB, Kang L, Song Q, Fan Z, Maloney WJ, Wang Y. Cryptotanshinone Protects Cartilage against Developing Osteoarthritis through the miR-106a-5p/GLIS3 Axis. MOLECULAR THERAPY. NUCLEIC ACIDS 2018; 11:170-179. [PMID: 29858052 PMCID: PMC5992348 DOI: 10.1016/j.omtn.2018.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 02/05/2018] [Accepted: 02/05/2018] [Indexed: 02/06/2023]
Abstract
Cryptotanshinone (CTS) has emerged as an anti-inflammatory agent in osteoarthritis (OA). However, the molecular mechanism underlying its potent therapeutic effect on OA remains largely unknown. MicroRNAs (miRNAs) act as crucial regulators in maintaining cartilage homeostasis. To investigate whether CTS protects against developing OA through regulation of miRNAs, we examined the potential CTS-mediated miRNA molecules using microarray analysis. We found that CTS significantly promoted miR-106a-5p expression in chondrocytes. Using the OA mouse model created by anterior cruciate ligament transection, we revealed that intra-articular injection of miR-106a-5p agomir attenuated OA. In addition, miR-106a-5p inhibited GLI-similar 3 (GLIS3) production by directly targeting the 3′ untranslated region. CTS promoted miR-106a-5p expression through recruitment of a member of the paired box (PAX) family of transcription factors, PAX5, to the miR-106a-5p promoter. Inhibition of PAX5 mimicked the effect of miR-106a-5p and abolished the CTS ability to regulate miR-106a-5p expression. In OA patients, miR-106-5p is downregulated which is accompanied by downregulation of PAX5 and upregulation of GLIS3. Collectively, these data highlight that the PAX5/miR-106a-5p/GLIS3 axis acts as a novel pleiotropic regulator in CTS-mediated OA cartilage protection, suggesting that miR-106a-5p and PAX5 activation and GLIS3 inhibition might be useful and attractive for therapeutic strategies to treat OA patients.
Collapse
Affiliation(s)
- Quanbo Ji
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, China; Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Dengbin Qi
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Xiaojie Xu
- Department of Medical Molecular Biology, Beijing Institute of Biotechnology, Beijing 100850, China
| | - Yameng Xu
- Department of Traditional Chinese Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Stuart B Goodman
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Lei Kang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing 100034, China
| | - Qi Song
- Department of Oncology, General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Zhongyi Fan
- Department of Oncology, General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - William J Maloney
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA 94305, USA.
| | - Yan Wang
- Department of Orthopaedics, General Hospital of Chinese People's Liberation Army, Beijing 100853, China.
| |
Collapse
|
21
|
Li Y, Lu J, Bao X, Wang X, Wu J, Li X, Hong W. MiR-499-5p protects cardiomyocytes against ischaemic injury via anti-apoptosis by targeting PDCD4. Oncotarget 2018; 7:35607-35617. [PMID: 27231854 PMCID: PMC5094948 DOI: 10.18632/oncotarget.9597] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 05/17/2016] [Indexed: 12/18/2022] Open
Abstract
Recent studies have reported that miRNAs might play critical roles in acute myocardial infarction (AMI). The objective of this study is to investigate the role of miR-499-5p in AMI and its potential molecular mechanisms. The expression level of MiR-499-5p was remarkably decreased in the infarcted myocardial tissues and in the cultured neonatal rat cardiomyocytes induced by hypoxia. Overexpression or knockdown of miR-499-5p decreased or increased the apoptotic rates of cultured cardiomyocytes in vitro. In addition, ectopic overexpression of miR-499-5p in the rat AMI models with agomir reduced the myocardial infarct size through decreasing the cardiomyocytes apoptosis in the infarcted area of the rat hearts. PDCD4 (programmed cell death 4) was verified as a direct target of miR-499-5p by luciferase report assay, and ectopic overexpression or inhibition of miR-499-5p could inhibit or increase the PDCD4 expression at both the mRNA and protein levels. Furthermore, we found that ectopic overexpression of PDCD4 without miR-499-5p binding sites reversed miR-499-5p-mediated cardiomyocytes apoptosis. Together, these findings revealed the role of miR-499-5p in protecting the cardiomyocytes against apoptosis induced by AMI via its direct target PDCD4, which providing evidence for the miR-499-5p/PDCD4 pathway as a potential therapeutic target for patients with AMI.
Collapse
Affiliation(s)
- Yingqing Li
- Department of Emergency, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Jianhua Lu
- Department of Emergency, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xueming Bao
- Department of Emergency, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xifu Wang
- Department of Emergency, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Junhua Wu
- Department of Emergency, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Xiongbin Li
- Department of Emergency, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Weiqiang Hong
- Department of Emergency, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
22
|
Dong Y, Liu C, Zhao Y, Ponnusamy M, Li P, Wang K. Role of noncoding RNAs in regulation of cardiac cell death and cardiovascular diseases. Cell Mol Life Sci 2018; 75:291-300. [PMID: 28913665 PMCID: PMC11105653 DOI: 10.1007/s00018-017-2640-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/17/2017] [Accepted: 08/31/2017] [Indexed: 12/15/2022]
Abstract
Loss of functional cardiomyocytes is a major underlying mechanism for myocardial remodeling and heart diseases, due to the limited regenerative capacity of adult myocardium. Apoptosis, programmed necrosis, and autophagy contribute to loss of cardiac myocytes that control the balance of cardiac cell death and cell survival through multiple intricate signaling pathways. In recent years, non-coding RNAs (ncRNAs) have received much attention to uncover their roles in cell death of cardiovascular diseases, such as myocardial infarction, cardiac hypertrophy, and heart failure. In addition, based on the view that mitochondrial morphology is linked to three types of cell death, ncRNAs are able to regulate mitochondrial fission/fusion of cardiomyocytes by targeting genes involved in cell death pathways. This review focuses on recent progress regarding the complex relationship between apoptosis/necrosis/autophagy and ncRNAs in the context of myocardial cell death in response to stress. This review also provides insight into the treatment for heart diseases that will guide novel therapies in the future.
Collapse
Affiliation(s)
- Yanhan Dong
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao, 266021, China
| | - Cuiyun Liu
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao, 266021, China
| | - Yanfang Zhao
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao, 266021, China
| | - Murugavel Ponnusamy
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao, 266021, China
| | - Peifeng Li
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao, 266021, China.
| | - Kun Wang
- Institute for Translational Medicine, Qingdao University, Deng Zhou Road 38, Qingdao, 266021, China.
| |
Collapse
|
23
|
Zhang Z, Li H, Chen S, Li Y, Cui Z, Ma J. Knockdown of MicroRNA-122 Protects H9c2 Cardiomyocytes from Hypoxia-Induced Apoptosis and Promotes Autophagy. Med Sci Monit 2017; 23:4284-4290. [PMID: 28871076 PMCID: PMC5597036 DOI: 10.12659/msm.902936] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute myocardial infarction (AMI) is a severe disease causing heart failure and sudden death. Studies indicate that microRNAs (miRNAs) are involved in the pathophysiology of AMI. In the present study, we carefully explored the effects of miR-122 on myocardial hypoxia injury and its possible underlying mechanism. MATERIAL AND METHODS miR-122 expression was analyzed in H9c2 cardiomyocytes after being transfected with miR-122 mimic, ASO-miR-122, or negative control. Cell viability and apoptosis were investigated by CCK-8 assays and flow cytometry analysis, respectively. Cell migration was analyzed using wound-healing assays. Western blotting was performed to analyze the expression of phosphatase and tensin homolog deleted on chromosome 10 (PTEN)/phosphatidylinositol 3-hydroxy kinase (PI3K)/AKT and LC3-II/LC3-I. RESULTS Hypoxia exposure significantly inhibited H9c2 cell viability (P<0.01). miR-122 overexpression promoted the hypoxia-induced H9c2 cell proliferation and migration loss (P<0.05), and cell apoptosis was increased (P<0.05). miR-122 knockdown enhanced cell viability and decreased cell apoptosis (P<0.05). Knockdown of miR-122 enhanced PTEN/PI3K/AKT activation and cell autophagy. Overexpression of miR-122 inhibited the PTEN/PI3K/AKT pathway and cell autophagy pathway. CONCLUSIONS The expression of miR-122 is involved in hypoxia-induced H9c2 cardiomyocyte injury. Knockdown of miR-122 protects H9c2 cells from hypoxia-induced apoptosis and enhances cell viability.
Collapse
Affiliation(s)
- Zaiwei Zhang
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Hu Li
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Shasha Chen
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Ying Li
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Zhiyuan Cui
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| | - Jie Ma
- Department of Cardiovascular Medicine, Jining No.1 People's Hospital, Jining, Shandong, China (mainland)
| |
Collapse
|
24
|
Chen X, Zhao W, Yuan Y, Bai Y, Sun Y, Zhu W, Du Z. MicroRNAs tend to synergistically control expression of genes encoding extensively-expressed proteins in humans. PeerJ 2017; 5:e3682. [PMID: 28828274 PMCID: PMC5560240 DOI: 10.7717/peerj.3682] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 07/22/2017] [Indexed: 01/02/2023] Open
Abstract
Considering complicated microRNA (miRNA) biogenesis and action mechanisms, it was thought so high energy-consuming for a cell to afford simultaneous over-expression of many miRNAs. Thus it prompts that an alternative miRNA regulation pattern on protein-encoding genes must exist, which has characteristics of energy-saving and precise protein output. In this study, expression tendency of proteins encoded by miRNAs’ target genes was evaluated in human organ scale, followed by quantitative assessment of miRNA synergism. Expression tendency analysis suggests that universally expressed proteins (UEPs) tend to physically interact in clusters and participate in fundamental biological activities whereas disorderly expressed proteins (DEPs) are inclined to relatively independently execute organ-specific functions. Consistent with this, miRNAs that mainly target UEP-encoding mRNAs, such as miR-21, tend to collaboratively or even synergistically act with other miRNAs in fine-tuning protein output. Synergistic gene regulation may maximize miRNAs’ efficiency with less dependence on miRNAs’ abundance and overcome the deficiency that targeting plenty of genes by single miRNA makes miRNA-mediated regulation high-throughput but insufficient due to target gene dilution effect. Furthermore, our in vitro experiment verified that merely 25 nM transfection of miR-21 be sufficient to influence the overall state of various human cells. Thus miR-21 was identified as a hub in synergistic miRNA–miRNA interaction network. Our findings suggest that synergistic miRNA–miRNA interaction is an important endogenous miRNA regulation mode, which ensures adequate potency of miRNAs at low abundance, especially those implicated in fundamental biological regulation.
Collapse
Affiliation(s)
- Xue Chen
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University (Institute of Clinical Pharmacy, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University), Harbin, China
| | - Wei Zhao
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University (Institute of Clinical Pharmacy, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University), Harbin, China
| | - Ye Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University (Institute of Clinical Pharmacy, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University), Harbin, China
| | - Yan Bai
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University (Institute of Clinical Pharmacy, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University), Harbin, China
| | - Yong Sun
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University (Institute of Clinical Pharmacy, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University), Harbin, China
| | - Wenliang Zhu
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University (Institute of Clinical Pharmacy, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University), Harbin, China
| | - Zhimin Du
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University (Institute of Clinical Pharmacy, The Heilongjiang Key Laboratory of Drug Research, Harbin Medical University), Harbin, China
| |
Collapse
|
25
|
Long B, Gan TY, Zhang RC, Zhang YH. miR-23a Regulates Cardiomyocyte Apoptosis by Targeting Manganese Superoxide Dismutase. Mol Cells 2017; 40:542-549. [PMID: 28756653 PMCID: PMC5582300 DOI: 10.14348/molcells.2017.0012] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 06/22/2017] [Accepted: 06/25/2017] [Indexed: 02/06/2023] Open
Abstract
Cardiomyocyte apoptosis is initiated by various cellular insults and accumulated cardiomyocyte apoptosis leads to the pathogenesis of heart failure. Excessive reactive oxygen species (ROS) provoke apoptotic cascades. Manganese superoxide dismutase (MnSOD) is an important antioxidant enzyme that converts cellular ROS into harmless products. In this study, we demonstrate that MnSOD is down-regulated upon hydrogen peroxide treatment or ischemia/reperfusion (I/R) injury. Enhanced expression of MnSOD attenuates cardiomyocyte apoptosis and myocardial infarction induced by I/R injury. Further, we show that miR-23a directly regulates the expression of MnSOD. miR-23a regulates cardiomyocyte apoptosis by suppressing the expression of MnSOD. Our study reveals a novel model regulating cardiomyocyte apoptosis which is composed of miR-23a and MnSOD. Our study provides a new method to tackling apoptosis related cardiac diseases.
Collapse
Affiliation(s)
- Bo Long
- Central Research Laboratory, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730,
China
| | - Tian-Yi Gan
- State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037,
China
| | - Rong-Cheng Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037,
China
| | - Yu-Hui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure center, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing 100037,
China
| |
Collapse
|
26
|
Fu BC, Lang JL, Zhang DY, Sun L, Chen W, Liu W, Liu KY, Ma CY, Jiang SL, Li RK, Tian H. Suppression of miR-34a Expression in the Myocardium Protects Against Ischemia-Reperfusion Injury Through SIRT1 Protective Pathway. Stem Cells Dev 2017; 26:1270-1282. [PMID: 28599575 DOI: 10.1089/scd.2017.0062] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
MicroRNA-34a (miR-34a) is expressed in the myocardium and expression is altered after myocardial injury. We investigated the effects of miR-34a on heart function after ischemia-reperfusion (IR) injury. Cardiomyocytes were isolated from neonatal rat hearts and simulated IR injury was induced in vitro. Following IR injury in rats, infarct size was measured and left ventricular (LV) function was evaluated using echocardiography. Protein expression of silent information regulator 1 (SIRT1), acetylated p53 (ac-p53), Bcl-2 and Bax, and miR-34a and SIRT1 gene levels were analyzed. miR-34a overexpression exacerbated myocardial injury by increasing apoptosis and infarct size and decreasing LV function. Suppression of miR-34a attenuated myocardial IR injury. SIRT1 was negatively regulated by miR-34a and the expression of downstream genes, such as ac-p53, Bcl-2, and Bax were altered correspondingly. Increased expression of miR-34a aggravates injury after IR; miR-34a suppression therapy may represent a new line of treatment for myocardial IR injury.
Collapse
Affiliation(s)
- Bi-Cheng Fu
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Ji-Lu Lang
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Dong-Yang Zhang
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Lu Sun
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Wei Chen
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Wei Liu
- 2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Kai-Yu Liu
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Chong-Yi Ma
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Shu-Lin Jiang
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| | - Ren-Ke Li
- 3 Division of Cardiovascular Surgery, Toronto General Research Institute, University Health Network , Toronto, Canada .,4 Division of Cardiac Surgery, Department of Surgery, University of Toronto , Toronto, Canada
| | - Hai Tian
- 1 Department of Cardiovascular Surgery, The Second Affiliated Hospital of Harbin Medical University , Harbin, China .,2 Key Laboratories of Myocardial Ischemia, Chinese Ministry of Education , Harbin, China
| |
Collapse
|
27
|
Gao CK, Liu H, Cui CJ, Liang ZG, Yao H, Tian Y. Roles of MicroRNA-195 in cardiomyocyte apoptosis induced by myocardial ischemia-reperfusion injury. J Genet 2016; 95:99-108. [PMID: 27019437 DOI: 10.1007/s12041-016-0616-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study aims to investigate microRNA-195 (miR-195) expression in myocardial ischaemia-reperfusion (I/R) injury and the roles of miR-195 in cardiomyocyte apoptosis though targeting Bcl-2. A mouse model of I/R injury was established. MiR- 195 expression levels were detected by real-time quantitative PCR (qPCR), and the cardiomyocyte apoptosis was detected by TUNEL assay. After cardiomyocytes isolated from neonatal rats and transfected with miR-195 mimic or inhibitor, the hypoxia/reoxygenation (H/R) injury model was established. Cardiomyocyte apoptosis and mitochondrial membrane potential were evaluated using flow cytometry. Bcl-2 and Bax mRNA expressions were detected by RT-PCR. Bcl-2, Bax and cytochrome c (Cyt-c) protein levels were determined by Western blot. Caspase-3 and caspase-9 activities were assessed by luciferase assay. Compared with the sham group, miR-195 expression levels and rate of cardiomyocyte apoptosis increased significantly in I/R group (both P < 0.05). Compared to H/R + negative control (NC) group, rate of cardiomyocyte apoptosis increased in H/R + miR-195 mimic group while decreased in H/R + miR-195 inhibitor group (both P <0.05). MiR-195 knockdown alleviated the loss of mitochondrial membrane potential (P <0.05). MiR-195 overexpression decreased Bcl-2 mRNA and protein expression, increased BaxmRNA and protein expression, Cyt-c protein expression and caspase-3 and caspase-9 activities (all P <0.05).While, downregulated MiR-195 increased Bcl-2 mRNA and protein expression, decreased Bax mRNA and protein expression, Cyt-c protein expression and caspase-3 and caspase-9 activities (all P < 0.05). Our study identified that miR-195 expression was upregulated in myocardial I/R injury, and miR-195 overexpression may promote cardiomyocyte apoptosis by targeting Bcl-2 and inducing mitochondrial apoptotic pathway.
Collapse
Affiliation(s)
- Chang-Kui Gao
- Department of Emergency, Longnan Hospital of Daqing, Daqing 163001, People's Republic of
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
Autophagy is a self-digestive process regulated by an intricate network of factors able either to ensure the prosurvival function of autophagy or to convert it in a death pathway. Recently, the involvement of miRNAs in the regulation of autophagy networks has been reported. This review will summarize the main features of these small noncoding endogenous RNAs, focusing on their relevance in cancer and finally addressing their impact on autophagy.
Collapse
|
29
|
Habibi P, Alihemmati A, Nasirzadeh M, Yousefi H, Habibi M, Ahmadiasl N. Involvement of microRNA-133 and -29 in cardiac disturbances in diabetic ovariectomized rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2016; 19:1177-1185. [PMID: 27917273 PMCID: PMC5126218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
OBJECTIVES Menopause and diabetes obviously increase the risk of cardiovascular disease in women. The aims of the present study were to evaluate the effects of ovariectomy in type 2 diabetes on the histology and expression of miRNA-29, miRNA-133, IGF-1 and Bcl-2 genes and Bcl-2 protein and caspase 3 activity in the hearts of female rats. MATERIALS AND METHODS Forty Female Wistar rats were divided into four groups: control, sham, ovariectomized (OVX), and ovariectomized with type 2 diabetes (OVX.D). After the 8-week experiment, the histological evaluation of the heart tissue was performed using H&E staining and PAS analysis, and cardiac expression of miRNA-29, miRNA-133, IGF-1, and Bcl-2 were evaluated using real-time PCR, and Bcl-2 protein and caspase 3 activity were evaluated using Western blot and ELISA. RESULTS Ovariectomy significantly decreased miRNA-29, miRNA-133, IGF-1, and BCL-2 expression and Bcl-2 protein and increased caspase 3 activity in the heart compared to sham animals group (P<0.05). Type 2 diabetes in ovariectomized rats markedly decreased expression of miRNA-29, miRNA-133, IGF-1, BCL-2 genes, and Bcl-2 protein, and increased caspase 3 activity and reduced collagen and fibroblast tissue and glycogen granule deposition in relation to OVX group (P<0.05). CONCLUSION Our findings suggest that type 2 diabetes and menopause synergically could enhance the cardiac fibrosis through dysregulation of miRNA-29, miRNA-133, IGF-1, and Bcl-2 genes expression and Bcl-2 protein and upregulation of caspase 3 activity.
Collapse
Affiliation(s)
- Parisa Habibi
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Alihemmati
- Department of Histology & Embryology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hadi Yousefi
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Nasser Ahmadiasl
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran,Corresponding author: Nasser Ahmadiasl. Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran. Tel/Fax: +98-4133364664;
| |
Collapse
|
30
|
Vegter EL, Schmitter D, Hagemeijer Y, Ovchinnikova ES, van der Harst P, Teerlink JR, O'Connor CM, Metra M, Davison BA, Bloomfield D, Cotter G, Cleland JG, Givertz MM, Ponikowski P, van Veldhuisen DJ, van der Meer P, Berezikov E, Voors AA, Khan MAF. Use of biomarkers to establish potential role and function of circulating microRNAs in acute heart failure. Int J Cardiol 2016; 224:231-239. [PMID: 27661412 DOI: 10.1016/j.ijcard.2016.09.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Circulating microRNAs (miRNAs) emerge as potential heart failure biomarkers. We aimed to identify associations between acute heart failure (AHF)-specific circulating miRNAs and well-known heart failure biomarkers. METHODS Associations between 16 biomarkers predictive for 180day mortality and the levels of 12 AHF-specific miRNAs were determined in 100 hospitalized AHF patients, at baseline and 48hours. Patients were divided in 4 pre-defined groups, based on clinical parameters during hospitalization. Correlation analyses between miRNAs and biomarkers were performed and complemented by miRNA target prediction and pathway analysis. RESULTS No significant correlations were found at hospital admission. However, after 48hours, 7 miRNAs were significantly negatively correlated to biomarkers indicative for a worse clinical outcome in the patient group with the most unfavorable in-hospital course (n=21); miR-16-5p was correlated to C-reactive protein (R=-0.66, p-value=0.0027), miR-106a-5p to creatinine (R=-0.68, p-value=0.002), miR-223-3p to growth differentiation factor 15 (R=-0.69, p-value=0.0015), miR-652-3p to soluble ST-2 (R=-0.77, p-value<0.001), miR-199a-3p to procalcitonin (R=-0.72, p-value<0.001) and galectin-3 (R=-0.73, p-value<0.001) and miR-18a-5p to procalcitonin (R=-0.68, p-value=0.002). MiRNA target prediction and pathway analysis identified several pathways related to cardiac diseases, which could be linked to some of the miRNA-biomarker correlations. CONCLUSIONS The majority of correlations between circulating AHF-specific miRNAs were related to biomarkers predictive for a worse clinical outcome in a subgroup of worsening heart failure patients at 48hours of hospitalization. The selective findings suggest a time-dependent effect of circulating miRNAs and highlight the susceptibility to individual patient characteristics influencing potential relations between miRNAs and biomarkers.
Collapse
Affiliation(s)
- Eline L Vegter
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Daniela Schmitter
- Momentum Research, Inc., Hagmattstrasse 17, CH-4123 Allschwil, Switzerland
| | - Yanick Hagemeijer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Ekaterina S Ovchinnikova
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands; European Research Institute for the Biology of Ageing and University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - John R Teerlink
- University of California at San Francisco, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | | | - Marco Metra
- Cardiology, The Department of Medical and Surgical Specialties, Radiological Sciences, and Public Health, University of Brescia, Italy
| | | | | | | | - John G Cleland
- National Heart & Lung Institute, Royal Brompton & Harefield Hospitals, Imperial College, London, UK
| | - Michael M Givertz
- Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Dirk J van Veldhuisen
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Eugene Berezikov
- European Research Institute for the Biology of Ageing and University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Adriaan A Voors
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Mohsin A F Khan
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| |
Collapse
|
31
|
Wu BL, Wang D, Bai WJ, Zhang F, Zhao X, Yi Y, Zhang T, Shen WJ, Li EM, Xu LY, Xu JZ. An integrative framework to identify cell death-related microRNAs in esophageal squamous cell carcinoma. Oncotarget 2016; 7:56758-56766. [PMID: 27462775 PMCID: PMC5302951 DOI: 10.18632/oncotarget.10779] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Accepted: 07/06/2016] [Indexed: 02/05/2023] Open
Abstract
Cell death is a critical biological process involved in many important functions, and defects in this system are usually linked with numerous human diseases including cancers. Esophageal squamous cell carcinoma is one of the most chemo- and biological therapy resistant cancers. Based on knowledge repository and four miRNAs profiling data, we proposed a general framework to hunt for cell death miRNAs in a context dependent manner. We predicted 12 candidate miRNAs from hundreds of others. Follow-up experimental verification of 7 miRNAs indicated at least 3 miRNAs (MIR20b, MIR498 and MIR196) were involved in both apoptosis and autophagy processes. These results indicated miRNAs intimately connected the two cell death modules in esophageal squamous cell carcinoma. This integrative framework can also be easily extended to identify miRNAs in other key cellular signaling pathways or may find conditional specific miRNAs in other cancer types.
Collapse
Affiliation(s)
- Bing-Li Wu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Dong Wang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Wen-Jing Bai
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Fan Zhang
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China
| | - Xing Zhao
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China
| | - Ying Yi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Ting Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Wen-Jun Shen
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou 515041, China
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou 515041, China
| | - Jian-Zhen Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou 515041, China
- Computational Systems Biology Laboratory, Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30602, USA
- Department of Bioinformatics, Shantou University Medical College, Shantou 515041, China
| |
Collapse
|
32
|
Chu H, Kohane DS, Langer R. RNA therapeutics - The potential treatment for myocardial infarction. Regen Ther 2016; 4:83-91. [PMID: 31245491 PMCID: PMC6581817 DOI: 10.1016/j.reth.2016.03.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/05/2016] [Accepted: 03/05/2016] [Indexed: 01/19/2023] Open
Abstract
RNA therapeutics mainly control gene expression at the transcript level. In contrast to conventional gene therapy which solely increases production of a protein, delivered RNAs can enhance, reduce or abolish synthesis of a particular protein, which control its relevant activities in a more diverse fashion. Thus, they hold promise to treat many human diseases including myocardial infarction (MI). MI is a serious health burden that causes substantial morbidity and mortality. An unmet clinical need for treating MI is the recovery of cardiac function, which requires regeneration of the functional tissues including the vasculature, nerves, and myocardium. Several classes of RNA therapeutics have been investigated in preclinical MI models, and the results have demonstrated their benefits and encourage their future development. In this review, we summarize the common RNA therapeutic approaches and highlight their application in MI therapy.
Collapse
Affiliation(s)
- Hunghao Chu
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Daniel S. Kohane
- Laboratory for Biomaterials and Drug Delivery, Department of Anesthesiology, Division of Critical Care Medicine, Children's Hospital Boston, Harvard Medical School, Boston, MA 02115, United States
| | - Robert Langer
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| |
Collapse
|
33
|
C-reactive protein levels in the perioperative period as a predictive marker of endoscopic recurrence after ileo-colonic resection for Crohn's disease. Cell Death Discov 2016; 2:16032. [PMID: 27551522 PMCID: PMC4979416 DOI: 10.1038/cddiscovery.2016.32] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/11/2016] [Accepted: 04/15/2016] [Indexed: 12/22/2022] Open
Abstract
The aim of this study was to determine the perioperative behavior of C-reactive protein (CRP) in Crohn’s disease (CD) patients undergoing elective ileo-cecal (IC) resection and to identify association between perioperative CRP levels and endoscopic recurrence at 1 year. Study hypothesis was that perioperative CRP changes are disease specific and could detect subset of patients with more aggressive pathopysiology. Seventy-five patients undergoing IC resection for CD were prospectively enrolled. Serial CRP levels were assessed: preoperative, postoperative day 1 (POD1) and day 5 (POD5). CD patients’ values were compared against same interval assessments of control groups undergoing right colectomy and appendicectomy. At POD1, the serum concentration increase was significantly higher in CD patients than in controls. Comparing with control groups, CRP levels remained remarkably high and showed a lower reduction in CD at POD5. Difference between groups was statistically significant. Optimal cutoff levels have been identified: serum CRP concentrations of >39.8 mg/l at POD1 and of >23.2 mg/l at POD5 have shown a significant association to endoscopic recurrence when using bivariate correlation. In this preliminary series, binary logistic regression could not demonstrate statistical relationship between endoscopic recurrence and any of the variables evaluated as prognostic factor. This is the only study so far that investigates and confirms a disease-specific upregulation of CRP response in the perioperative period for CD patients undergoing surgery. The postoperative CRP levels and kinetics seem to be related to the grade of mucosal inflammation and recurrence rate according to our 12 months endoscopic evaluation.
Collapse
|
34
|
Bernardo BC, Nguyen SS, Gao XM, Tham YK, Ooi JYY, Patterson NL, Kiriazis H, Su Y, Thomas CJ, Lin RCY, Du XJ, McMullen JR. Inhibition of miR-154 Protects Against Cardiac Dysfunction and Fibrosis in a Mouse Model of Pressure Overload. Sci Rep 2016; 6:22442. [PMID: 26928825 PMCID: PMC4772383 DOI: 10.1038/srep22442] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/15/2016] [Indexed: 01/19/2023] Open
Abstract
Expression of miR-154 is upregulated in the diseased heart and was previously shown to be upregulated in the lungs of patients with pulmonary fibrosis. However, the role of miR-154 in a model of sustained pressure overload-induced cardiac hypertrophy and fibrosis had not been assessed. To examine the role of miR-154 in the diseased heart, adult male mice were subjected to transverse aortic constriction for four weeks, and echocardiography was performed to confirm left ventricular hypertrophy and cardiac dysfunction. Mice were then subcutaneously administered a locked nucleic acid antimiR-154 or control over three consecutive days (25 mg/kg/day) and cardiac function was assessed 8 weeks later. Here, we demonstrate that therapeutic inhibition of miR-154 in mice with pathological hypertrophy was able to protect against cardiac dysfunction and attenuate adverse cardiac remodelling. The improved cardiac phenotype was associated with attenuation of heart and cardiomyocyte size, less cardiac fibrosis, lower expression of atrial and B-type natriuretic peptide genes, attenuation of profibrotic markers, and increased expression of p15 (a miR-154 target and cell cycle inhibitor). In summary, this study suggests that miR-154 may represent a novel target for the treatment of cardiac pathologies associated with cardiac fibrosis, hypertrophy and dysfunction.
Collapse
Affiliation(s)
| | - Sally S Nguyen
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia.,Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, 3086, Australia
| | - Xiao-Ming Gao
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
| | - Yow Keat Tham
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia.,Monash University, Clayton, 3800, Australia
| | - Jenny Y Y Ooi
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
| | | | - Helen Kiriazis
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
| | - Yidan Su
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia
| | - Colleen J Thomas
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, 3086, Australia
| | - Ruby C Y Lin
- Asbestos Diseases Research Institute, Concorde Hospital, 2139, Australia.,Ramaciotti Centre for Genomics and School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, 2052, Australia
| | - Xiao-Jun Du
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia.,Monash University, Clayton, 3800, Australia
| | - Julie R McMullen
- Baker IDI Heart and Diabetes Institute, Melbourne, 3004, Australia.,Monash University, Clayton, 3800, Australia
| |
Collapse
|
35
|
Abstract
Diabetes mellitus is a metabolic homeostasis disease that contributes to additional comorbidities such as cardiovascular disease (CVD) and cancer. It has a long undiagnosed latent period during which there can be irreparable damage to the pancreas and cardiovascular tissues. Recent studies have highlighted the roles of several microRNAs in CVD. Determining the microRNAs that link diabetes mellitus and CVD is an important topic to be explored. In the present review, we discuss the microRNAs that contribute to the progression of diabetes mellitus and CVD and focus on the miR-29 family microRNAs whose expression is upregulated by hyperglycemia and proinflammatory cytokines, the hallmarks of diabetes mellitus. Upregulation of miR-29 expression is a key factor in the loss of pancreatic β cells and development of the first stage of type 1 diabetes mellitus (T1DM). Additionally, miR-29-mediated suppression of myeloid cell leukemia 1 (MCL-1), an important prosurvival protein, underlies Marfan's syndrome, abdominal aortic aneurysm, and diabetes mellitus-associated cardiomyocyte disorganization. Suppression of miR-29 expression and subsequent increase in the prosurvival MCL-1, however, promotes tumor development. Therefore, miR-29 mimics that suppress MCL-1 are hailed as tumor suppressors. The critical question is whether an increase in miR-29 levels is well tolerated in conditions of comorbidities in which insulin resistance is an underlying disease. In light of increasing awareness of the interconnection of diabetes mellitus, CVD, and cancer, it is of utmost importance to understand the mechanism of action of current treatment options on all of the comorbidities and careful evaluation of cardiovascular toxicity must accompany any treatment paradigm that increases miR-29 levels.
Collapse
Affiliation(s)
- Anna Ślusarz
- aDepartment of Medicine bDepartment of Biochemistry, University of Missouri cHarry S. Truman Memorial Veterans Affairs Hospital dDepartment of Nutrition and Exercise Physiology, University of Missouri, Columbia, Missouri, USA
| | | |
Collapse
|
36
|
Lu J, Gu H, Tang Q, Wu W, Yuan B, Guo D, Wei Y, Sun H, Xia Y, Ding H, Hu L, Chen D, Sha J, Wang X. Common SNP in hsa-miR-196a-2 increases hsa-miR-196a-5p expression and predisposes to idiopathic male infertility in Chinese Han population. Sci Rep 2016; 6:19825. [PMID: 26805933 PMCID: PMC4726409 DOI: 10.1038/srep19825] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 12/07/2015] [Indexed: 01/05/2023] Open
Abstract
MicroRNA plays an important role in spermatogenesis. Whether pre-miRNAs polymorphisms are associated with idiopathic male infertility remains obscure. In this study, 1378 idiopathic infertile males and 486 fertile controls were included between 2006 and 2014. Genotype of three polymorphisms (hsa-mir-146a rs2910164, hsa-mir-196a-2 rs11614913, and hsa-mir-499 rs3746444) and expression of miRNA in seminal plasma were examined by TaqMan method. The role of hsa-miR-196a-5p in cell proliferation, apoptosis and cell cycle were also examined in GC-2 cells. Our results demonstrated that rs11614913 of hsa-miR-196a-2 was significantly associated with idiopathic infertility (TT vs. CT: P = 0.014; TT vs. CC: P = 0.005; TT vs. CT + CC: P = 0.003). In following stratified analysis, we found that rs11614913 exhibited a significantly higher risk of asthenospermia, oligozoospermia and azoospermia. However, no significant association was observed between the other two polymorphisms and idiopathic male infertility risk. In a genotype-expression correlation analysis, rs11614913 CC was significantly associated with elevated expression of hsa-miR-196a-5p (P < 0.05). Additionally, apoptosis levels were significantly increased in hsa-miR-196a-5p mimic treated GC-2 cells, while decreased in hsa-miR-196a-5p inhibitor treated GC-2 cells. Our data revealed a significant relationship between hsa-miR-196a-2 polymorphism and idiopathic male infertility.
Collapse
Affiliation(s)
- Jing Lu
- State Key Laboratory of Reproductive Medicine, Department of Reproduction, Nanjing Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Hao Gu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 210029, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Qiuqin Tang
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics, Nanjing Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 210029, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, China.,Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Beilei Yuan
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 210029, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Dan Guo
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 210029, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Yongyue Wei
- Department of Epidemiology and Biostatistics, Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, 210029, China
| | - Hong Sun
- Department of Microbial and Molecular Systems, KULeuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 210029, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| | - Hongjuan Ding
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics, Nanjing Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Lingqing Hu
- Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Daozhen Chen
- Wuxi Maternal and Child Health Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Jiahao Sha
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 210029, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
37
|
Jin C, Yan B, Lu Q, Lin Y, Ma L. Reciprocal regulation of Hsa-miR-1 and long noncoding RNA MALAT1 promotes triple-negative breast cancer development. Tumour Biol 2015; 37:7383-94. [PMID: 26676637 DOI: 10.1007/s13277-015-4605-6] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 12/07/2015] [Indexed: 02/06/2023] Open
Abstract
Recent studies demonstrated that long noncoding RNAs (lncRNAs) have a critical role in the regulation of cancer progression and metastasis. However, little is known about the mechanism through which metastasis-associated lung adencarcinoma transcript 1 (MALAT1) exerts its oncogenic activity, and the interaction between MALAT1 and microRNA remains largely unknown. In the present study, we reported that MALAT1 was upregulated in triple-negative breast cancer (TNBC) tissues. Knockdown of MALAT1 inhibited proliferation, motility, and increased apoptosis in vitro. In vivo study indicated that knockdown of MALAT1 inhibited tumor growth and metastasis. Patients with high MALAT1 expression had poorer overall survival time than those with low MALAT1 expression. In addition, our findings demonstrate a reciprocal negative control relationship between MALAT1 and miR-1: downregulation of MALAT1 increased expression of microRNA-1 (miR-1), while overexpression of miR-1 decreased MALAT1 expression. Slug was identified as a direct target of miR-1. We proposed that MALAT1 exerted its function through the miR-1/slug axis. In summary, we proposed that MALAT1 may be a target for TNBC therapy.
Collapse
MESH Headings
- 3' Untranslated Regions/genetics
- Animals
- Cell Line, Tumor
- DNA, Recombinant/genetics
- Female
- Gene Expression Regulation, Neoplastic
- Humans
- Kaplan-Meier Estimate
- Mice, Inbred BALB C
- Mice, Nude
- MicroRNAs/biosynthesis
- MicroRNAs/genetics
- Middle Aged
- Neoplasm Invasiveness
- Neoplasm Proteins/biosynthesis
- Neoplasm Proteins/genetics
- Neoplasm Proteins/physiology
- RNA/genetics
- RNA Interference
- RNA, Long Noncoding/biosynthesis
- RNA, Long Noncoding/genetics
- RNA, Neoplasm/biosynthesis
- RNA, Neoplasm/genetics
- RNA, Small Interfering/genetics
- Snail Family Transcription Factors/biosynthesis
- Snail Family Transcription Factors/genetics
- Snail Family Transcription Factors/physiology
- Specific Pathogen-Free Organisms
- Triple Negative Breast Neoplasms/genetics
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/mortality
- Triple Negative Breast Neoplasms/pathology
Collapse
Affiliation(s)
- Chuan Jin
- The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, China
| | - Bingchuan Yan
- The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, China
| | - Qin Lu
- The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yanmin Lin
- Oncology Center, The Affiliated Hospital of Guangdong Medical College, Zhanjiang, China
| | - Lei Ma
- The Affiliated Cancer Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
38
|
Gurianova V, Stroy D, Ciccocioppo R, Gasparova I, Petrovic D, Soucek M, Dosenko V, Kruzliak P. Stress response factors as hub-regulators of microRNA biogenesis: implication to the diseased heart. Cell Biochem Funct 2015; 33:509-18. [PMID: 26659949 DOI: 10.1002/cbf.3151] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 09/21/2015] [Accepted: 10/02/2015] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNAs) are important regulators of heart function and then an intriguing therapeutic target for plenty of diseases. The problem raised is that many data in this area are contradictory, thus limiting the use of miRNA-based therapy. The goal of this review is to describe the hub-mechanisms regulating the biogenesis and function of miRNAs, which could help in clarifying some contradictions in the miRNA world. With this scope, we analyse an array of factors, including several known agents of stress response, mediators of epigenetic changes, regulators of alternative splicing, RNA editing, protein synthesis and folding and proteolytic systems. All these factors are important in cardiovascular function and most of them regulate miRNA biogenesis, but their influence on miRNAs was shown for non-cardiac cells or some specific cardiac pathologies. Finally, we consider that studying the stress response factors, which are upstream regulators of miRNA biogenesis, in the diseased heart could help in (1) explaining some contradictions concerning miRNAs in heart pathology, (2) making the role of miRNAs in pathogenesis of cardiovascular disease more clear, and therefore, (3) getting powerful targets for its molecular therapy.
Collapse
Affiliation(s)
- Veronika Gurianova
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Dmytro Stroy
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Rachele Ciccocioppo
- Clinica Medica I; Fondazione IRCCS Policlinico San Matteo, Università degli Studi di Pavia, Italy
| | - Iveta Gasparova
- Institute of Biology, Genetics and Medical Genetics, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovak Republic
| | - Daniel Petrovic
- Institute of Histology and Embryology, Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Miroslav Soucek
- Second Department of Internal Medicine, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic
| | - Victor Dosenko
- Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kiev, Ukraine
| | - Peter Kruzliak
- Second Department of Internal Medicine, St. Anne's University Hospital and Masaryk University, Brno, Czech Republic.,Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University, Bratislava, Slovak Republic.,Laboratory of Structural Biology and Proteomics, Faculty of Pharmacy, Faculty of Pharmacy, University of Veterinary and Pharmaceutical Sciences, Brno, Czech Republic
| |
Collapse
|
39
|
Mukhadi S, Hull R, Mbita Z, Dlamini Z. The Role of MicroRNAs in Kidney Disease. Noncoding RNA 2015; 1:192-221. [PMID: 29861424 PMCID: PMC5932548 DOI: 10.3390/ncrna1030192] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 09/28/2015] [Accepted: 11/08/2015] [Indexed: 02/07/2023] Open
Abstract
MicroRNAs (miRNAs) are short noncoding RNAs that regulate pathophysiological processes that suppress gene expression by binding to messenger RNAs. These biomolecules can be used to study gene regulation and protein expression, which will allow better understanding of many biological processes such as cell cycle progression and apoptosis that control the fate of cells. Several pathways have also been implicated to be involved in kidney diseases such as Transforming Growth Factor-β, Mitogen-Activated Protein Kinase signaling, and Wnt signaling pathways. The discovery of miRNAs has provided new insights into kidney pathologies and may provide new innovative and effective therapeutic strategies. Research has demonstrated the role of miRNAs in a variety of kidney diseases including renal cell carcinoma, diabetic nephropathy, nephritic syndrome, renal fibrosis, lupus nephritis and acute pyelonephritis. MiRNAs are implicated as playing a role in these diseases due to their role in apoptosis, cell proliferation, differentiation and development. As miRNAs have been detected in a stable condition in different biological fluids, they have the potential to be tools to study the pathogenesis of human diseases with a great potential to be used in disease prognosis and diagnosis. The purpose of this review is to examine the role of miRNA in kidney disease.
Collapse
Affiliation(s)
- Sydwell Mukhadi
- Forensic Science Laboratory, 730 Pretorius street, Arcadia 0083, South Africa.
| | - Rodney Hull
- College of Agriculture and Environmental Sciences, University of South Africa, Private Bag X6, Florida 1709, Johannesburg 1709, South Africa.
| | - Zukile Mbita
- Department of Biochemistry, Microbiology and Biotechnology, University of Limpopo, Private Bag x1106, Sovenga 0727, South Africa.
| | - Zodwa Dlamini
- Research, Innovation & Engagements Portfolio, Mangosuthu University of Technology, Durban 4031, South Africa.
| |
Collapse
|
40
|
miRNA therapeutics: a new class of drugs with potential therapeutic applications in the heart. Future Med Chem 2015; 7:1771-92. [PMID: 26399457 DOI: 10.4155/fmc.15.107] [Citation(s) in RCA: 181] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
miRNAs are small non-coding RNAs (ncRNAs), which regulate gene expression. Here, the authors describe the contribution of miRNAs to cardiac biology and disease. They discuss various strategies for manipulating miRNA activity including antisense oligonucleotides (antimiRs, blockmiRs), mimics, miRNA sponges, Tough Decoys and miRNA mowers. They review developments in chemistries (e.g., locked nucleic acid) and modifications (sugar, 'ZEN', peptide nucleic acids) and miRNA delivery tools (viral vectors, liposomes, nanoparticles, pHLIP). They summarize potential miRNA therapeutic targets for heart disease based on preclinical studies. Finally, the authors review current progress of miRNA therapeutics in clinical development for HCV and cancer, and discuss challenges that will need to be overcome for similar therapies to enter the clinic for patients with cardiac disease.
Collapse
|
41
|
|
42
|
Yuan D, Cui X, Wang Y, Zhao Y, Li H, Hu S, Chu X, Li Y, Li Q, Liu Q, Zhu W. Enrichment Analysis Identifies Functional MicroRNA-Disease Associations in Humans. PLoS One 2015; 10:e0136285. [PMID: 26296081 PMCID: PMC4546424 DOI: 10.1371/journal.pone.0136285] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Accepted: 08/02/2015] [Indexed: 11/18/2022] Open
Abstract
Substantial evidence has shown that microRNAs (miRNAs) may be causally linked to the occurrence and progression of human diseases. Herein, we conducted an enrichment analysis to identify potential functional miRNA-disease associations (MDAs) in humans by integrating currently known biological data: miRNA-target interactions (MTIs), protein-protein interactions, and gene-disease associations. Two contributing factors to functional miRNA-disease associations were quantitatively considered: the direct effects of miRNA that target disease-related genes, and indirect effects triggered by protein-protein interactions. Ninety-nine miRNAs were scanned for possible functional association with 2223 MeSH-defined human diseases. Each miRNA was experimentally validated to target ≥ 10 mRNA genes. Putative MDAs were identified when at least one MTI was confidently validated for a disease. Overall, 19648 putative MDAs were found, of which 10.0% was experimentally validated. Further results suggest that filtering for miRNAs that target a greater number of disease-related genes (n ≥ 8) can significantly enrich for true MDAs from the set of putative associations (enrichment rate = 60.7%, adjusted hypergeometric p = 2.41×10-91). Considering the indirect effects of miRNAs further elevated the enrichment rate to 72.6%. By using this method, a novel MDA between miR-24 and ovarian cancer was found. Compared with scramble miRNA overexpression of miR-24 was validated to remarkably induce ovarian cancer cells apoptosis. Our study provides novel insight into factors contributing to functional MDAs by integrating large quantities of previously generated biological data, and establishes a feasible method to identify plausible associations with high confidence.
Collapse
Affiliation(s)
- Dandan Yuan
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaomeng Cui
- School of Measurement and Control Technology & Communications Engineering, Harbin University of Science and Technology, Harbin, China
| | - Yang Wang
- Department of Pharmacy, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yilei Zhao
- Department of Pharmacy, the First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huiying Li
- Department of Obstetrics and Gynecology, Hongqi Hospital of Mudanjiang Medical University, Mudanjiang, China
| | - Suangjiu Hu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaodan Chu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yan Li
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qiang Li
- Department of General Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (QL); (WZ)
| | - Wenliang Zhu
- Institute of Clinical Pharmacology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
- * E-mail: (QL); (WZ)
| |
Collapse
|
43
|
Liu R, Li J, Lai Y, Liao Y, Liu R, Qiu W. Hsa-miR-1 suppresses breast cancer development by down-regulating K-ras and long non-coding RNA MALAT1. Int J Biol Macromol 2015; 81:491-7. [PMID: 26275461 DOI: 10.1016/j.ijbiomac.2015.08.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 07/11/2015] [Accepted: 08/07/2015] [Indexed: 12/31/2022]
Abstract
MicroRNAs exert their functions by mainly regulating coding genes or long non-coding RNA expression. In the present study, we reported that hsa-miR-1 was down-regulated in breast cancer tissues. Restoration of miR-1 in breast cancer cells inhibited proliferation, motility and increased apoptosis in vitro. MiR-1 functioned as a tumor suppressor by targeting K-RAS and MALAT1. In addition, the effects of up-regulation of miR-1 were similar to that of silencing K-RAS and MALAT1 in breast cancer cells. In vivo study indicated that restoration of miR-1 inhibited tumor growth and metastasis. Patients with low miR-1 expression had poorer overall survival time than those with high miR-1 expression. Our findings emphasized the potential role of miR-1 as tumor suppressive miRNA in breast cancer.
Collapse
Affiliation(s)
- Ruilei Liu
- Department of Thyroid & Breast Surgery, The Third Affiliated Hospital of Sun Yet-sen University, Guangzhou, China
| | - Jie Li
- Department of Thyroid & Breast Surgery, The First Affiliated Hospital of Sun Yet-sen University, Guangzhou, China
| | - Yuanhui Lai
- Department of Vascular & Thyroid & Breast Surgery, The First Affiliated Hospital of Sun Yet-sen University, Guangzhou, China
| | - Yi Liao
- Department of Gastrointestinal Surgery, The Sixth Affiliated Hospital of Sun Yet-sen University, Guangzhou, China
| | - Ruiming Liu
- Laboratory of Department of Surgery, The First Affiliated Hospital of Sun Yet-sen University, Guangzhou, China
| | - Wanshou Qiu
- Department of Thyroid & Breast Surgery, The Third Affiliated Hospital of Sun Yet-sen University, Guangzhou, China.
| |
Collapse
|
44
|
Parikh A, Wu J, Blanton RM, Tzanakakis ES. Signaling Pathways and Gene Regulatory Networks in Cardiomyocyte Differentiation. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:377-92. [PMID: 25813860 DOI: 10.1089/ten.teb.2014.0662] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Strategies for harnessing stem cells as a source to treat cell loss in heart disease are the subject of intense research. Human pluripotent stem cells (hPSCs) can be expanded extensively in vitro and therefore can potentially provide sufficient quantities of patient-specific differentiated cardiomyocytes. Although multiple stimuli direct heart development, the differentiation process is driven in large part by signaling activity. The engineering of hPSCs to heart cell progeny has extensively relied on establishing proper combinations of soluble signals, which target genetic programs thereby inducing cardiomyocyte specification. Pertinent differentiation strategies have relied as a template on the development of embryonic heart in multiple model organisms. Here, information on the regulation of cardiomyocyte development from in vivo genetic and embryological studies is critically reviewed. A fresh interpretation is provided of in vivo and in vitro data on signaling pathways and gene regulatory networks (GRNs) underlying cardiopoiesis. The state-of-the-art understanding of signaling pathways and GRNs presented here can inform the design and optimization of methods for the engineering of tissues for heart therapies.
Collapse
Affiliation(s)
- Abhirath Parikh
- 1 Lonza Walkersville, Inc. , Lonza Group, Walkersville, Maryland
| | - Jincheng Wu
- 2 Department of Chemical and Biological Engineering, Tufts University , Medford, Massachusetts
| | - Robert M Blanton
- 3 Division of Cardiology, Molecular Cardiology Research Institute , Tufts Medical Center, Tufts School of Medicine, Boston, Massachusetts
| | - Emmanuel S Tzanakakis
- 2 Department of Chemical and Biological Engineering, Tufts University , Medford, Massachusetts.,4 Tufts Clinical and Translational Science Institute (CTSI) , Boston, Massachusetts
| |
Collapse
|
45
|
Slezak J, Kura B, Ravingerová T, Tribulova N, Okruhlicova L, Barancik M. Mechanisms of cardiac radiation injury and potential preventive approaches. Can J Physiol Pharmacol 2015; 93:737-53. [PMID: 26030720 DOI: 10.1139/cjpp-2015-0006] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In addition to cytostatic treatment and surgery, the most common cancer treatment is gamma radiation. Despite sophisticated radiological techniques however, in addition to irradiation of the tumor, irradiation of the surrounding healthy tissue also takes place, which results in various side-effects, depending on the absorbed dose of radiation. Radiation either damages the cell DNA directly, or indirectly via the formation of oxygen radicals that in addition to the DNA damage, react with all cell organelles and interfere with their molecular mechanisms. The main features of radiation injury besides DNA damage is inflammation and increased expression of pro-inflammatory genes and cytokines. Endothelial damage and dysfunction of capillaries and small blood vessels plays a particularly important role in radiation injury. This review is focused on summarizing the currently available data concerning the mechanisms of radiation injury, as well as the effectiveness of various antioxidants, anti-inflammatory cytokines, and cytoprotective substances that may be utilized in preventing, mitigating, or treating the toxic effects of ionizing radiation on the heart.
Collapse
Affiliation(s)
- Jan Slezak
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Branislav Kura
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Táňa Ravingerová
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Narcisa Tribulova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Ludmila Okruhlicova
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| | - Miroslav Barancik
- Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic.,Institute for Heart Research, Slovak Academy of Sciences, Dúbravská cesta 9, 842 33 Bratislava, Slovak Republic
| |
Collapse
|