1
|
Cheng K, Zhai Q, Song J, Liu B. The Co-pathogenic Target Gene CNTN1 Involved in Coronary Artery Disease and Pulmonary Arterial Hypertension Has Potential for Diagnosis of Coronary Artery Disease. Anatol J Cardiol 2024; 28:381-392. [PMID: 39087405 PMCID: PMC11317787 DOI: 10.14744/anatoljcardiol.2024.4331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/22/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND We aimed to find a gene for coronary artery disease (CAD) early diagnosis by detecting co-pathogenic target gene involved in CAD and pulmonary arterial hypertension (PAH).
Methods: Datasets were obtained from the Gene Expression Omnibus (GEO) database, including GSE113079, GSE113439, and GSE12288, to investigate gene expression patterns in cardiovascular diseases. Weighted Gene Co-expression Network Analysis (WGCNA) was performed to identify gene modules associated with clinical traits. Differential gene expression analysis and functional enrichment analysis were carried out. Protein-protein interaction (PPI) networks were constructed. JASPAR database and FIMO tool were utilized to predict transcription factor (TF) binding sites.
Results: Fifteen key genes were identified in CAD and PAH, with CNTN1 being prioritized for further investigation due to its high connectivity degree. Upstream regulation analysis identified potential TFs (DRGX, HOXD3, and RAX) and 7 miRNAs targeting CNTN1. The expression profile of CNTN1 was significantly upregulated in CAD samples, and ROC analysis indicated potential diagnostic value for CAD. CMap database analysis predicted potential targeted drugs for CAD.
Conclusion: CNTN1 was detected as a co-pathogenetic gene for CAD and PAH. It is highly expressed in CAD patients and has potential value for CAD diagnosis. CNTN1 is potentially regulated by 3 TFs and 7 miRNAs.
Collapse
Affiliation(s)
- Kun Cheng
- Department of Cardiovascular Surgery, Zibo Central Hospital, Shandong Province, China
| | - Qixuan Zhai
- Department of Cardiovascular Surgery, Zibo Central Hospital, Shandong Province, China
| | - Jieqiong Song
- Department of Blood Transfusion, Zibo Central Hospital, Shandong Province, China
| | - Bing Liu
- Department of Cardiovascular Surgery, Zibo Central Hospital, Shandong Province, China
| |
Collapse
|
2
|
Yang J, Zhai Y, Huang C, Xiang Z, Liu H, Wu J, Huang Y, Liu L, Li W, Wang W, Yang J, Zhang J. RP105 Attenuates Ischemia/Reperfusion-Induced Oxidative Stress in the Myocardium via Activation of the Lyn/Syk/STAT3 Signaling Pathway. Inflammation 2024; 47:1371-1385. [PMID: 38568415 DOI: 10.1007/s10753-024-01982-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/10/2024] [Accepted: 01/22/2024] [Indexed: 08/24/2024]
Abstract
Although our previous studies have established the crucial role of RP105 in myocardial ischemia/reperfusion injury (MI/RI), its involvement in regulating oxidative stress induced by MI/RI remains unclear. To investigate this, we conducted experiments using a rat model of ischemia/reperfusion (I/R) injury. Adenovirus carrying RP105 was injected apically at multiple points, and after 72 h, the left anterior descending coronary artery was ligated for 30 min followed by 2 h of reperfusion. In vitro experiments were performed on H9C2 cells, which were transfected with recombinant adenoviral vectors for 48 h, subjected to 4 h of hypoxia, and then reoxygenated for 2 h. We measured oxidative stress markers, including superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) activities, as well as malondialdehyde (MDA) concentration, using a microplate reader. The fluorescence intensity of reactive oxygen species (ROS) in myocardial tissue was measured using a DHE probe. We also investigated the upstream and downstream components of the signal transducer and activator of transcription 3 (STAT3). Upregulation of RP105 increased SOD and GSH-Px activities, reduced MDA concentration, and inhibited ROS production in response to I/R injury in vivo and hypoxia reoxygenation (H/R) stimulation in vitro. The overexpression of RP105 led to a decrease in the myocardial enzyme LDH in serum and cell culture supernatant, as well as a reduction in infarct size. Additionally, left ventricular fraction (LVEF) and fractional shortening (LVFS) were improved in the RP105 overexpression group compared to the control. Upregulation of RP105 promoted the expression of Lyn and Syk and further activated STAT phosphorylation, which was blocked by PP2 (a Lyn inhibitor). Our findings suggest that RP105 can inhibit MI/RI-induced oxidative stress by activating STAT3 via the Lyn/Syk signaling pathway.
Collapse
Affiliation(s)
- Jian Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Yuhong Zhai
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Cuiyuan Huang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Zujin Xiang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Haiyin Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Jingyi Wu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Yifan Huang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Li Liu
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Wenqiang Li
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Wei Wang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China.
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yiling Road 183, Yichang, 443000, Hubei, China.
- Hubei Key Laboratory of Ischemic Cardiovascular Disease, Yichang, 443000, China.
- HuBei Clinical Research Center for Ischemic Cardiovascular Disease, Yichang, 443000, China.
| |
Collapse
|
3
|
Huang K, Yang W, Shi M, Wang S, Li Y, Xu Z. The Role of TPM3 in Protecting Cardiomyocyte from Hypoxia-Induced Injury via Cytoskeleton Stabilization. Int J Mol Sci 2024; 25:6797. [PMID: 38928503 PMCID: PMC11203979 DOI: 10.3390/ijms25126797] [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: 05/12/2024] [Revised: 06/10/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Ischemic heart disease (IHD) remains a major global health concern, with ischemia-reperfusion injury exacerbating myocardial damage despite therapeutic interventions. In this study, we investigated the role of tropomyosin 3 (TPM3) in protecting cardiomyocytes against hypoxia-induced injury and oxidative stress. Using the AC16 and H9c2 cell lines, we established a chemical hypoxia model by treating cells with cobalt chloride (CoCl2) to simulate low-oxygen conditions. We found that CoCl2 treatment significantly upregulated the expression of hypoxia-inducible factor 1 alpha (HIF-1α) in cardiomyocytes, indicating the successful induction of hypoxia. Subsequent morphological and biochemical analyses revealed that hypoxia altered cardiomyocyte morphology disrupted the cytoskeleton, and caused cellular damage, accompanied by increased lactate dehydrogenase (LDH) release and malondialdehyde (MDA) levels, and decreased superoxide dismutase (SOD) activity, indicative of oxidative stress. Lentivirus-mediated TPM3 overexpression attenuated hypoxia-induced morphological changes, cellular damage, and oxidative stress imbalance, while TPM3 knockdown exacerbated these effects. Furthermore, treatment with the HDAC1 inhibitor MGCD0103 partially reversed the exacerbation of hypoxia-induced injury caused by TPM3 knockdown. Protein-protein interaction (PPI) network and functional enrichment analysis suggested that TPM3 may modulate cardiac muscle development, contraction, and adrenergic signaling pathways. In conclusion, our findings highlight the therapeutic potential of TPM3 modulation in mitigating hypoxia-associated cardiac injury, suggesting a promising avenue for the treatment of ischemic heart disease and other hypoxia-related cardiac pathologies.
Collapse
Affiliation(s)
- Ke Huang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China;
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Weijia Yang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Mingxuan Shi
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Shiqi Wang
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Yi Li
- Key Laboratory of Dental Maxillofacial Reconstruction and Biological Intelligence Manufacturing, School of Stomatology, Lanzhou University, Lanzhou 730030, China; (W.Y.); (M.S.); (S.W.)
| | - Zhaoqing Xu
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730030, China;
| |
Collapse
|
4
|
Jiang Y, Liu Z, Ye L, Cheng J, Wan J. MiR-449b-5p Ameliorates Hypoxia-induced Cardiomyocyte Injury through Activating PI3K/AKT Pathway by Targeting BCL2L13. Appl Biochem Biotechnol 2024:10.1007/s12010-024-04931-5. [PMID: 38581629 DOI: 10.1007/s12010-024-04931-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2024] [Indexed: 04/08/2024]
Abstract
Recent reports show miR-449b-5p reduces liver and renal ischemia/reperfusion (I/R) injury, but its effects on hypoxia-induced cardiomyocyte injury in ischemic heart disease are still unknown. In this study, AC16 human cardiomyocytes underwent hypoxic conditions for durations of 24, 48, and 72 h. We observed that miR-449b-5p expression was significantly downregulated in hypoxic AC16 cardiomyocytes. Elevating the levels of miR-449b-5p in these cells resulted in enhanced cell survival, diminished release of LDH, and a reduction in cell apoptosis and oxidative stress using CCK-8, LDH assays, flow cytometry, TUNEL staining, and various commercial kits. Conversely, reducing miR-449b-5p levels resulted in the opposite effects. Through bioinformatics analysis and luciferase reporter assays, BCL2-like 13 (BCL2L13) was determined to be a direct target of miR-449b-5p. Inhibiting BCL2L13 greatly inhibited hypoxia-induced cell viability loss, LDH release, cell apoptosis, and excessive production of oxidative stress, whereas increasing BCL2L13 negated miR-449b-5p's protective impact in hypoxic AC16 cardiomyocytes. Additionally, miR-449b-5p elevated the levels of the proteins p-PI3K, p-AKT, and Bcl-2, while decreasing Bax expression in hypoxic AC16 cardiomyocytes by targeting BCL2L13. In summary, the research indicates that the protective effects of miR-449b-5p are facilitated through the activation of the PI3K/AKT pathway, which promotes cell survival, and by targeting BCL2L13, which inhibits apoptosis, offering a potential therapeutic strategy for ischemic heart disease by mitigating hypoxia-induced cardiomyocyte injury.
Collapse
Affiliation(s)
- Yang Jiang
- Department of Emergency Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, Anhui, 230000, China
| | - Zeyan Liu
- Department of Emergency Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, Anhui, 230000, China
| | - Li Ye
- Department of Emergency Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, Anhui, 230000, China
| | - Jinglin Cheng
- Department of Emergency Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, Anhui, 230000, China
| | - Jun Wan
- Department of Emergency Medicine, The Second Affiliated Hospital of Anhui Medical University, No. 678 Furong Road, Jingkai District, Hefei, Anhui, 230000, China.
| |
Collapse
|
5
|
Zhan Y, Li L, Guo C, Zhang Y, Zhao L, Tao Z, Zhang H, Chen S. MicroRNA-141-3p reduces pulmonary hypoxia/reoxygenation injury through suppression of Beclin-1-dependent autophagy. Aging (Albany NY) 2024; 16:1352-1373. [PMID: 38261732 PMCID: PMC10866419 DOI: 10.18632/aging.205430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/06/2023] [Indexed: 01/25/2024]
Abstract
Alterations in autophagy are involved in pulmonary hypoxia/reoxygenation (H/R)-induced injury. Here, we intended to explain the function of microRNA-141-3p (miR-141-3p) in regulating autophagy under the H/R condition. Rat pulmonary microvascular endothelial cells (PMVECs) were applied for H/R cell model establishment, followed by tracing of autophagy formation. SIRT1 plays a critical role in controlling the lifespan of yeast, flies, and mice. Interaction between SIRT1 and Beclin-1, an indicator protein for autophagy, and between miR-141-3p and SIRT1 was assayed with their roles in PMVEC injury. Autophagy of PMVECs was activated after hypoxia treatment and further activated after H/R treatment. The binding of miR-141-3p and SIRT1 was verified. In H/R-treated PMVECs, the binding of miR-141-3p and SIRT1 was reduced. Furthermore, SIRT1 acted as a deacetylase to stabilize the Beclin-1 protein, promoting autophagy and PMVEC injury. H/R rat models were established, and in vivo, experiments further confirmed that miR-141-3p regulated autophagy and lung injury in H/R rats through SIRT1/Beclin-1 axis. The current study highlighted that reduced miR-141-3p in H/R-treated PMVECs promoted deacetylation of Beclin-1 by SIRT1, thus causing PMVEC injury.
Collapse
Affiliation(s)
- Yanping Zhan
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Lei Li
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Chen Guo
- Jiangxi Maternal and Child Health Hospital, Nanchang 330006, P.R. China
| | - Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Lili Zhao
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Zhe Tao
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| | - Hua Zhang
- Nanchang University, Nanchang 330006, P.R. China
| | - Shibiao Chen
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, P.R. China
| |
Collapse
|
6
|
Gehris J, Ervin C, Hawkins C, Womack S, Churillo AM, Doyle J, Sinusas AJ, Spinale FG. Fibroblast activation protein: Pivoting cancer/chemotherapeutic insight towards heart failure. Biochem Pharmacol 2024; 219:115914. [PMID: 37956895 PMCID: PMC10824141 DOI: 10.1016/j.bcp.2023.115914] [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: 08/25/2023] [Revised: 11/06/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
An important mechanism for cancer progression is degradation of the extracellular matrix (ECM) which is accompanied by the emergence and proliferation of an activated fibroblast, termed the cancer associated fibroblast (CAF). More specifically, an enzyme pathway identified to be amplified with local cancer progression and proliferation of the CAF, is fibroblast activation protein (FAP). The development and progression of heart failure (HF) irrespective of the etiology is associated with left ventricular (LV) remodeling and changes in ECM structure and function. As with cancer, HF progression is associated with a change in LV myocardial fibroblast growth and function, and expresses a protein signature not dissimilar to the CAF. The overall goal of this review is to put forward the postulate that scientific discoveries regarding FAP in cancer as well as the development of specific chemotherapeutics could be pivoted to target the emergence of FAP in the activated fibroblast subtype and thus hold translationally relevant diagnostic and therapeutic targets in HF.
Collapse
Affiliation(s)
- John Gehris
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlie Ervin
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Charlotte Hawkins
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Sydney Womack
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Amelia M Churillo
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Jonathan Doyle
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States
| | - Albert J Sinusas
- Yale University Cardiovascular Imaging Center, New Haven CT, United States
| | - Francis G Spinale
- Cell Biology and Anatomy and Cardiovascular Research Center, University of South Carolina School of Medicine and the Columbia VA Health Care System, Columbia, SC, United States.
| |
Collapse
|
7
|
Hu T, Li D, Fan T, Zhao X, Chen Z. CircCRIM1/microRNA-141-3p/thioredoxin-binding protein axis mediates neuronal apoptosis after cerebral ischemia-reperfusion. ENVIRONMENTAL TOXICOLOGY 2023; 38:2845-2856. [PMID: 37565716 DOI: 10.1002/tox.23916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/06/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
Numerous studies have indicated enrichment of circular RNA (circRNA) in the brain takes on a momentous role in cerebral ischemia-reperfusion (CIR) injury. A recent study discovered a novel circCRIM1, was highly expressed in the middle cerebral artery occlusion-reperfusion (MCAO/R) model. Nevertheless, its specific biological function remained unknown. The study was to explore circCRIM1 in CIR-induced neuronal apoptosis. As measured, circCRIM1 and TXNIP were up-regulated, while miR-141-3p was down-regulated in MCAO/R mouse model and OGD/R SH-SY5Y cells. Depleting circCRIM1 reduced the number of apoptotic neurons in MCAO/R rats, increased the number of Nissl bodies, prevented reactive oxygen species production and oxidative stress imbalance in brain tissues, repressed cleaved caspase-3, Bax, and Cyto C protein levels and increased Bcl-2 levels. Overexpression of circCRIM1 further repressed neuronal activity and accelerated apoptosis in OGD/R model, disrupted redox balance. Depleting circCRIM1 had the opposite effect in OGD/R model. Knocking down miR-141-3p or TXNIP weakened the effects of knocking down circCRIM1 or overexpressing circCRIM1, separately. Mechanistically, circCRIM1 exerted an active role in CIR injury via miR-141-3p to mediate TXNIP. All in all, the circCRIM1/miR-141-3p/TXNIP axis might be a latent therapeutic target for CIR injury.
Collapse
Affiliation(s)
- Teng Hu
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - Di Li
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - TiePing Fan
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - XuSheng Zhao
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| | - ZhongJun Chen
- Department of Neurological Intervention, Dalian Municipal Central Hospital, Dalian City, China
| |
Collapse
|
8
|
Edwards K, Lydyard PM, Kulikova N, Tsertsvadze T, Volpi EV, Chiorazzi N, Porakishvili N. The role of CD180 in hematological malignancies and inflammatory disorders. Mol Med 2023; 29:97. [PMID: 37460961 PMCID: PMC10353253 DOI: 10.1186/s10020-023-00682-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 06/08/2023] [Indexed: 07/20/2023] Open
Abstract
Toll-like receptors play a significant role in the innate immune system and are also involved in the pathophysiology of many different diseases. Over the past 35 years, there have been a growing number of publications exploring the role of the orphan toll-like receptor, CD180. We therefore set out to provide a narrative review of the current evidence surrounding CD180 in both health and disease. We first explore the evidence surrounding the role of CD180 in physiology including its expression, function and signaling in antigen presenting cells (APCs) (dendritic cells, monocytes, and B cells). We particularly focus on the role of CD180 as a modulator of other TLRs including TLR2, TLR4, and TLR9. We then discuss the role of CD180 in inflammatory and autoimmune diseases, as well as in hematological malignancies of B cell origin, including chronic lymphocytic leukemia (CLL). Based on this evidence we produce a current model for CD180 in disease and explore the potential role for CD180 as both a prognostic biomarker and therapeutic target. Throughout, we highlight specific areas of research which should be addressed to further the understanding of CD180 biology and the translational potential of research into CD180 in various diseases.
Collapse
Affiliation(s)
- Kurtis Edwards
- School of Life Sciences, University of Westminster, London, UK
| | - Peter M Lydyard
- School of Life Sciences, University of Westminster, London, UK.
- The University of Georgia, Tbilisi, Georgia.
- Division of Infection of Immunity, University College London, Gower Street, London, WC1E 6BT, UK.
| | - Nino Kulikova
- Agricultural University of Georgia, Tbilisi, Georgia
| | | | | | | | | |
Collapse
|
9
|
Silva Grijó Farani P, Iandra da Silva Ferreira B, Begum K, Vilar-Pereira G, Pereira IR, Fernández-Figueroa EA, Cardenas-Ovando RA, Almeida IC, Roy S, Lannes-Vieira J, Moreira OC. Treatment with benznidazole and pentoxifylline regulates microRNA transcriptomic profile in a murine model of Chagas chronic cardiomyopathy. PLoS Negl Trop Dis 2023; 17:e0011223. [PMID: 36972298 PMCID: PMC10121046 DOI: 10.1371/journal.pntd.0011223] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 04/21/2023] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
Chronic Chagas cardiomyopathy (CCC) is one of the leading causes of morbidity and mortality due to cardiovascular disorders in endemic areas of Chagas disease (CD), a neglected tropical illness caused by the protozoan parasite Trypanosoma cruzi. CCC is characterized by parasite persistence and inflammatory response in the heart tissue, which occur parallel to microRNA (miRNA) alterations. Here, we investigated the miRNA transcriptome profiling in the cardiac tissue of chronically T. cruzi-infected mice treated with a suboptimal dose of benznidazole (Bz), the immunomodulator pentoxifylline alone (PTX), or the combination of both (Bz+PTX), following the CCC onset. At 150 days post-infection, Bz, PTX, and Bz+PTX treatment regimens improved electrocardiographic alterations, reducing the percentage of mice afflicted by sinus arrhythmia and second-degree atrioventricular block (AVB2) when compared with the vehicle-treated animals. miRNA Transcriptome profiling revealed considerable changes in the differential expression of miRNAs in the Bz and Bz+PTX treatment groups compared with the control (infected, vehicle-treated) group. The latter showed pathways related to organismal abnormalities, cellular development, skeletal muscle development, cardiac enlargement, and fibrosis, likely associated with CCC. Bz-Treated mice exhibited 68 differentially expressed miRNAs related to signaling pathways like cell cycle, cell death and survival, tissue morphology, and connective tissue function. Finally, the Bz+PTX-treated group revealed 58 differentially expressed miRNAs associated with key signaling pathways related to cellular growth and proliferation, tissue development, cardiac fibrosis, damage, and necrosis/cell death. The T. cruzi-induced upregulation of miR-146b-5p, previously shown in acutely infected mice and in vitro T. cruzi-infected cardiomyocytes, was reversed upon Bz and Bz+PTX treatment regimens when further experimentally validated. Our results further our understanding of molecular pathways related to CCC progression and evaluation of treatment response. Moreover, the differentially expressed miRNAs may serve as drug targets, associated molecular therapy, or biomarkers of treatment outcomes.
Collapse
Affiliation(s)
- Priscila Silva Grijó Farani
- Real-Time PCR Platform RPT09A, Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Beatriz Iandra da Silva Ferreira
- Real-Time PCR Platform RPT09A, Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Khodeza Begum
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, United States of America
| | - Glaucia Vilar-Pereira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Isabela Resende Pereira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Edith A. Fernández-Figueroa
- Computational and Integrative Genomics, Instituto Nacional de Medicina Genómica, Arenal Tepepan, Mexico City, Mexico
| | | | - Igor C. Almeida
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, United States of America
| | - Sourav Roy
- Border Biomedical Research Center, Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, United States of America
| | - Joseli Lannes-Vieira
- Laboratory of Biology of the Interactions, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Otacilio Cruz Moreira
- Real-Time PCR Platform RPT09A, Laboratory of Molecular Virology and Parasitology, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
10
|
Zhang H, Wang Y, Wu K, Liu R, Wang H, Yao Y, Kvietys P, Rui T. miR‑141 impairs mitochondrial function in cardiomyocytes subjected to hypoxia/reoxygenation by targeting Sirt1 and MFN2. Exp Ther Med 2022; 24:763. [PMID: 36561976 PMCID: PMC9748642 DOI: 10.3892/etm.2022.11699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial oxidative stress and dysfunction are major pathogenic features of cardiac injury induced by ischemia/reperfusion (I/R). MicroRNA-141 (miR-141) has been implicated in the mitochondrial dysfunction in cell-based models of oxidant stress. Thus, the main aim of the present study was to systematically assess the role of miR-141 in cardiomyocyte injury induced by simulated I/R. The challenge of HL-1 cardiomyocytes with hypoxia/reoxygenation (H/R) decreased cell viability, which was also associated with an increase in miR-141 expression. The H/R-induced cell injury was mitigated by a miR-141 inhibitor and exacerbated by a miR-141 mimic. Furthermore, H/R induced mitochondrial superoxide production, dysfunction (decreased oxygen utilization and membrane depolarization), as well as ultrastructural damage. These mitochondrial effects were mitigated by a miR-141 inhibitor and intensified by a miR-141 mimic. Luciferase reporter assay, reverse transcription-quantitative PCR, and western blot analyses identified sirtuin-1 (Sirt1) and mitofusin-2 (MFN2) as targets of miR-141. The silencing of Sirt1 reduced the MFN2 cardiomyocyte levels and reversed the alleviating effects of miR-141 inhibitor on mitochondrial function during H/R. Collectively, these findings suggest that miR-141 functions as a causative agent in cardiomyocyte injury induced by I/R, primarily by interfering with two mitochondrial regulatory proteins, Sirt1 and MFN2.
Collapse
Affiliation(s)
- Hao Zhang
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yaqiao Wang
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Kehan Wu
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Runmin Liu
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Hao Wang
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Yongwei Yao
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China
| | - Peter Kvietys
- Department of Physiological Sciences, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Tao Rui
- Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu 212002, P.R. China,Critical Care Western, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada,Critical Illness Research, Lawson Health Research Institute, London, ON N6A 4G5, Canada,Departments of Medicine, Pathology and Laboratory Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada,Correspondence to: Dr Tao Rui, Division of Cardiology, Department of Medicine, The Affiliated People's Hospital of Jiangsu University, 8 Dianli Road, Zhenjiang, Jiangsu 212002, P.R. China
| |
Collapse
|
11
|
You C, Liu J, Qiu R, Xu L, Dai F, Ni Q, Qiu W. MiR-141 Modulates Bone Marrow Mesenchymal Stem Cells (BMSCs) Osteogenic/Adipogenic Differentiation Under Oxidative Stress. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BMSCs Osteogenic differentiation is beneficial to the construction of bone tissue engineering. Oxidative stress can affect BMSCs differentiation. MiR-141 regulates BMSCs proliferation. However, MiR-141’s role in BMSCs osteogenic/adipogenic differentiation under oxidative stress
is unclear. Mice BMSCs were assigned into control group; oxidative stress group; and si-MiR-141 group followed by detecting miR-141 level. After 14 days of osteogenesis or adipogenesis induction, RUNX2, OPN and FABP4 mRNA level was analyzed together with analysis of ROS and SOD content, ALP
activity and TGFβ/smad signaling protein level by Western blot. Under oxidative stress, MiR-141 was significantly upregulated and RUNX2 and OPN level was decreased, along with increased ROS content and FABP4 level, reduced SOD and ALP activity and expression of TGFβ1
and smad2 (P < 0.05). Transfection of MiR-141 siRNA into BMSCs under oxidative stress down-regulated MiR-141, significantly upregulated RUNX2 and OPN, reduced ROS, elevated SOD activity, downregulated FABP4, and increased ALP activity and TGFβ1 and smad2 expression (P
< 0.05). In conclusion, MiR-141 expression is increased in BMSCs under oxidative stress. Down-regulating MiR-141 improves the redox imbalance through TGFβ/smad signaling pathway, promotes osteogenic differentiation of BMSCs and inhibits differentiation to adipocytes.
Collapse
Affiliation(s)
- Chuanfei You
- Department of Orthopedics, Peoples Hospital of Siyang County, Suqian, Jiangsu, 223700, China
| | - Jun Liu
- Department of Orthopaedics, Second Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210009, China
| | - Ruoyu Qiu
- Department of Rheumatoid Immunity, Nanjing Gulou Hospital Group Suqian People’s Hospital, Suqian, Jiangsu, 223800, China
| | - Leijun Xu
- Department of Orthopedics, Peoples Hospital of Siyang County, Suqian, Jiangsu, 223700, China
| | - Furen Dai
- Department of Orthopedics, Peoples Hospital of Siyang County, Suqian, Jiangsu, 223700, China
| | - Qianzhao Ni
- Department of Orthopedics, Peoples Hospital of Siyang County, Suqian, Jiangsu, 223700, China
| | - Weisheng Qiu
- Department of Orthopedics, Peoples Hospital of Siyang County, Suqian, Jiangsu, 223700, China
| |
Collapse
|
12
|
Chen K, Jiang T, Zeng Q, Wang S, Li L. Bone Marrow Mesenchymal Stem Co-Culture Inhibited the Proliferation of Endometrial Cancer Cells by Upregulating miR-141-3p/Death Associated Protein Kinase 1 (DAPK1) Axis. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
MicroRNAs are involved in the biological processes of a variety of cancers. Here, we intend to assess the effect of BMSC-derived exosomal miR-141-3p on endometrial cancer (EC) cells. EC tumor and normal tissues were collected to measure miR-141-3p and death associated protein kinase
1 (DAPK1) expression. EC cells transfected with miR-141-3p mimic were cultured with BMSCs followed by measuring cell proliferation, apoptosis and migration. miR-141-3p and DAPK1 level was significantly decreased in EC tissues and cell lines. After miR-141-3p mimics transfection, miR-141-3p
and DAPK1 expression was increased significantly, along with inhibited cell proliferation, migration and invasion and increased cell apoptosis. Moreover, co-culture with BMSC decreased EC cells in vitro activity, and upregulated miR-141-3p and DAPK1 expression. In conclusion, miR-141-3p
and DAPK1 is reduced in EC and miR-141-3p overexpression inhibited EC cell biological behaviors, indicating that miR-141-3p might be a potential target for treatment of EC.
Collapse
Affiliation(s)
- Keming Chen
- Department of Obstetrics and Gynecology, The First People’s of Jingzhou, Jingzhou, Hubei, 434000, China
| | - Tao Jiang
- Department of Obstetrics and Gynecology, The First People’s of Jingzhou, Jingzhou, Hubei, 434000, China
| | - Qingsong Zeng
- Department of Obstetrics and Gynecology, The First People’s of Jingzhou, Jingzhou, Hubei, 434000, China
| | - Sufei Wang
- Department of Obstetrics and Gynecology, The First People’s of Jingzhou, Jingzhou, Hubei, 434000, China
| | - Li Li
- Department of Obstetrics and Gynecology, The First People’s of Jingzhou, Jingzhou, Hubei, 434000, China
| |
Collapse
|
13
|
Walkowski B, Kleibert M, Majka M, Wojciechowska M. Insight into the Role of the PI3K/Akt Pathway in Ischemic Injury and Post-Infarct Left Ventricular Remodeling in Normal and Diabetic Heart. Cells 2022; 11:cells11091553. [PMID: 35563860 PMCID: PMC9105930 DOI: 10.3390/cells11091553] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/28/2022] [Accepted: 05/03/2022] [Indexed: 02/07/2023] Open
Abstract
Despite the significant decline in mortality, cardiovascular diseases are still the leading cause of death worldwide. Among them, myocardial infarction (MI) seems to be the most important. A further decline in the death rate may be achieved by the introduction of molecularly targeted drugs. It seems that the components of the PI3K/Akt signaling pathway are good candidates for this. The PI3K/Akt pathway plays a key role in the regulation of the growth and survival of cells, such as cardiomyocytes. In addition, it has been shown that the activation of the PI3K/Akt pathway results in the alleviation of the negative post-infarct changes in the myocardium and is impaired in the state of diabetes. In this article, the role of this pathway was described in each step of ischemia and subsequent left ventricular remodeling. In addition, we point out the most promising substances which need more investigation before introduction into clinical practice. Moreover, we present the impact of diabetes and widely used cardiac and antidiabetic drugs on the PI3K/Akt pathway and discuss the molecular mechanism of its effects on myocardial ischemia and left ventricular remodeling.
Collapse
Affiliation(s)
- Bartosz Walkowski
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
| | - Marcin Kleibert
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Miłosz Majka
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Correspondence: (M.K.); (M.M.)
| | - Małgorzata Wojciechowska
- Laboratory of Centre for Preclinical Research, Department of Experimental and Clinical Physiology, Medical University of Warsaw, Banacha 1b, 02-097 Warsaw, Poland; (B.W.); (M.W.)
- Invasive Cardiology Unit, Independent Public Specialist Western Hospital John Paul II, Daleka 11, 05-825 Grodzisk Mazowiecki, Poland
| |
Collapse
|
14
|
Ma M, Liang SC, Diao KY, Wang Q, He Y. Mitofilin Mitigates Myocardial Damage in Acute Myocardial Infarction by Regulating Pyroptosis of Cardiomyocytes. Front Cardiovasc Med 2022; 9:823591. [PMID: 35586659 PMCID: PMC9108246 DOI: 10.3389/fcvm.2022.823591] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Background Acute myocardial infarction (AMI) can lead to sudden cardiac death after prolonged ischemia or heart failure (HF) and impaired left ventricular pump function. However, the underlying mechanism remains largely unknown. The purpose of this study was to investigate the role of mitofilin in alleviating AMI. Methods Recombinant adenoviral vectors for mitofilin overexpression or mitofilin knockdown were constructed, respectively. A mouse AMI model was established and the effect of mitofilin on myocardial pyroptosis was examined by detecting the lactate dehydrogenase (LDH) level and inflammatory factors. Moreover, a cellular model of AMI was established by treating cardiomyocytes with hypoxia/reoxygenation (H/R). An enzyme-linked immunosorbent assay (ELISA) and a western blot analysis were used to detect the effect of mitofilin knockdown on the expression of pyroptosis-related factors. Furthermore, the regulatory role of mitofilin in PI3K/AKT pathway was evaluated by the western blot and PI3K inhibitor. Results Mitofilin was downregulated in the heart tissue of the AMI mice and H/R induced cardiomyocytes. The overexpression of mitofilin significantly alleviated AMI and reduced pyroptosis-related factors. Meanwhile, in cardiomyocytes, mitofilin knockdown aggravated cellular damages by promoting pyroptosis. Further analysis showed that the anti-pyroptotic effect of mitofilin was dependent on the activation of the PI3K/AKT signaling pathway. Conclusions Our study suggests that mitofilin regulates pyroptosis through the PI3K/AKT signaling pathway in cardiomyocytes to ameliorate AMI, which may serve as a therapeutic strategy for the management of AMI.
Collapse
Affiliation(s)
- Min Ma
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
- Department of Cardiology, The Sixth People's Hospital of Chengdu, Chengdu, China
| | - Shi-chu Liang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| | - Kai-yue Diao
- Department of Radiology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Qin Wang
- Department of Cardiology, The Sixth People's Hospital of Chengdu, Chengdu, China
| | - Yong He
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Xin X, Duan L, Yang H, Yu H, Bao Y, Jia D, Wu N, Qiao Y. miR-141-3p regulates saturated fatty acid-induced cardiomyocyte apoptosis through Notch1/PTEN/AKT pathway via targeting PSEN1. ENVIRONMENTAL TOXICOLOGY 2022; 37:741-753. [PMID: 34897970 DOI: 10.1002/tox.23439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 11/30/2021] [Accepted: 12/05/2021] [Indexed: 06/14/2023]
Abstract
It has been reported that miR-141-3p levels are markedly upregulated in the cardiomyocytes of obese rats induced by a high-fat diet. However, the role of miR-141-3p in myocardial lipotoxicity remains elusive. In the present study, the role of miR-141-3p in lipotoxic injury of H9c2 cells induced by palmitic acid (PA) and its possible mechanisms were assessed. The results indicated that miR-141-3p was significantly upregulated in PA-induced cardiomyocytes. miR-141-3p inhibitor enhanced the cell viability, reduced the release of lactate dehydrogenase (LDH), creatine kinase-MB (CK-MB), and troponin I (CTN-I), decreased cell apoptosis rate, and repressed the activation of mitochondrial apoptosis pathway in PA-treated H9c2, whereas treatment with miR-141-3p mimics resulted in the opposite effects. Mechanistically, it was further revealed that miR-141-3p could specifically bind to presenilin 1 (PSEN1) 3'UTR, and upregulating miR-141-3p levels reduced the expression of PSEN1, thereby inhibiting the activation of the Notch1/PTEN/AKT pathway. Additionally, inhibition of Notch1/AKT signaling pathway by its inhibitor could abrogate the effect of miR-141-3p on mitochondrial-mediated apoptosis induced by PA. In conclusion, the present study demonstrates that miR-141-3p regulates saturated fatty acid-induced cardiomyocyte apoptosis through Notch1/PTEN/AKT pathway via targeting PSEN1, which gains a new insight into the mechanisms of myocardial lipotoxic injury.
Collapse
Affiliation(s)
- Xin Xin
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Lian Duan
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Huimin Yang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Hang Yu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Yandong Bao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Dalin Jia
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Nan Wu
- The Central Laboratory, The First Affiliated Hospital of China Medical University, Liaoning, China
| | - Ying Qiao
- The Central Laboratory, The First Affiliated Hospital of China Medical University, Liaoning, China
| |
Collapse
|
16
|
Fan Z, Pathak JL, Ge L. The Potential Role of RP105 in Regulation of Inflammation and Osteoclastogenesis During Inflammatory Diseases. Front Cell Dev Biol 2021; 9:713254. [PMID: 34414191 PMCID: PMC8369417 DOI: 10.3389/fcell.2021.713254] [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: 05/22/2021] [Accepted: 07/09/2021] [Indexed: 11/16/2022] Open
Abstract
Inflammatory diseases have a negative impact on bone homeostasis via exacerbated local and systemic inflammation. Bone resorbing osteoclasts are mainly derived from hematopoietic precursors and bone marrow monocytes. Induced osteoclastogenesis during inflammation, autoimmunity, metabolic diseases, and cancers is associated with bone loss and osteoporosis. Proinflammatory cytokines, pathogen-associated molecular patterns, or endogenous pathogenic factors induce osteoclastogenic differentiation by binding to the Toll-like receptor (TLR) family expressed on surface of osteoclast precursors. As a non-canonical member of the TLRs, radioprotective 105 kDa (RP105 or CD180) and its ligand, myeloid differentiation protein 1 (MD1), are involved in several bone metabolic disorders. Reports from literature had demonstrated RP105 as an important activator of B cells, bone marrow monocytes, and macrophages, which regulates inflammatory cytokines release from immune cells. Reports from literature had shown the association between RP105 and other TLRs, and the downstream signaling mechanisms of RP105 with different “signaling-competent” partners in immune cells during different disease conditions. This review is focused to summarize: (1) the role of RP105 on immune cells’ function and inflammation regulation (2) the potential regulatory roles of RP105 in different disease-mediated osteoclast activation and the underlying mechanisms, and (3) the different “signaling-competent” partners of RP105 that regulates osteoclastogenesis.
Collapse
Affiliation(s)
- Zhou Fan
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Janak L Pathak
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China
| | - Linhu Ge
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, China.,Institute of Oral Disease, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Rastgar Rezaei Y, Zarezadeh R, Nikanfar S, Oghbaei H, Nazdikbin N, Bahrami-Asl Z, Zarghami N, Ahmadi Y, Fattahi A, Nouri M, Dittrich R. microRNAs in the pathogenesis of non-obstructive azoospermia: the underlying mechanisms and therapeutic potentials. Syst Biol Reprod Med 2021; 67:337-353. [PMID: 34355990 DOI: 10.1080/19396368.2021.1951890] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
miRNAs are involved in different biological processes, including proliferation, differentiation, and apoptosis. Interestingly, 38% of the X chromosome-linked miRNAs are testis-specific and have crucial roles in regulating the renewal and cell cycle of spermatogonial stem cells. Previous studies demonstrated that abnormal expression of spermatogenesis-related miRNAs could lead to nonobstructive azoospermia (NOA). Moreover, differential miRNAs expression in seminal plasma of NOA patients has been reported compared to normozoospermic men. However, the role of miRNAs in NOA pathogenesis and the underlying mechanisms have not been comprehensively studied. Therefore, the aim of this review is to mechanistically describe the role of miRNAs in the pathogenesis of NOA and discuss the possibility of using the miRNAs as therapeutic targets.Abbreviations: AMO: anti-miRNA antisense oligonucleotide; AZF: azoospermia factor region; CDK: cyclin-dependent kinase; DAZ: deleted in azoospermia; ESCs: embryonic stem cells; FSH: follicle-stimulating hormone; ICSI: intracytoplasmic sperm injection; JAK/STAT: Janus kinase/signal transducers and activators of transcription; miRNA: micro-RNA; MLH1: Human mutL homolog l; NF-κB: Nuclear factor-kappa B; NOA: nonobstructive azoospermia; OA: obstructive azoospermia; PGCs: primordial germ cells; PI3K/AKT: Phosphatidylinositol 3-kinase/protein kinase B; Rb: retinoblastoma tumor suppressor; ROS: Reactive Oxygen Species; SCOS: Sertoli cell-only syndrome; SIRT: sirtuin; SNPs: single nucleotide polymorphisms; SSCs: spermatogonial stem cells; TESE: testicular sperm extraction; TGF-β: transforming growth factor-beta.
Collapse
Affiliation(s)
- Yeganeh Rastgar Rezaei
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saba Nikanfar
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hajar Oghbaei
- Department of Physiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Zahra Bahrami-Asl
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nosratollah Zarghami
- Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Ahmadi
- Department of Urology, Sina Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Fattahi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| | - Mohammad Nouri
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Medical Biotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ralf Dittrich
- Department of Obstetrics and Gynecology, Erlangen University Hospital, Friedrich-Alexander University of Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
Song W, Wang J, Zhang Y, Ma T, Wang K. Effect of Substance P on Differentiation of Bone Marrow Stromal Stem Cells Under Oxidative Stress. J BIOMATER TISS ENG 2021. [DOI: 10.1166/jbt.2021.2577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Bone marrow stromal stem cells (BMSCs) can be used to treat bone defects but BMSCs are damaged under oxidative stress. The neuropeptide substance P (SP) involves various cellular activities. However, SP’s role in BMSCs differentiation under oxidative stress is unknown. Rat BMSCs
were isolated and assigned into control group; oxidative stress group treated with 200 μM H2O2; and SP group, in which 10 mM SP was added under oxidative stress followed by analysis of SP secretion by ELISA, cell proliferation by MTT method, Caspase3 activity, Bax
and Bcl-2 level by Real time PCR, ALP activity ROS and SOD content as well as NF-κB level by Western blot. Under oxidative stress, SP secretion was significantly decreased, BMSCs proliferation was inhibited, Caspase3 activity and Bax expression increased, Bcl-2 and ALP activity was decreased
along with increased ROS activity and NF-κB level and reduced SOD activity (P <0.05), adding SP to BMSCs under oxidative stress can significantly promote SP secretion and cell proliferation, reduce Caspase3 activity and Bax expression, increase Bcl-2 expression and ALP activity,
decreased ROS activity and NF-κB level, and elevated SOD activity (P <0.05). SP secretion from BMSCs cells was reduced under oxidative stress. Up-regulation of SP in BMSCs cells under oxidative stress can inhibit BMSCs apoptosis and promote cell proliferation and osteogenesis
by regulating NF-κB.
Collapse
Affiliation(s)
- Wei Song
- First Department of Orthopedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shanxi, 710004, China
| | - Jun Wang
- Department of Joint Surgery, Hong-Hui Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shanxi, 710054, China
| | - Yumin Zhang
- Department of Joint Surgery, Hong-Hui Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shanxi, 710054, China
| | - Tao Ma
- Department of Joint Surgery, Hong-Hui Hospital, Xi’an Jiaotong University College of Medicine, Xi’an, Shanxi, 710054, China
| | - Kunzheng Wang
- First Department of Orthopedics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shanxi, 710004, China
| |
Collapse
|
19
|
Wu A, Sun W, Mou F. lncRNA‑MALAT1 promotes high glucose‑induced H9C2 cardiomyocyte pyroptosis by downregulating miR‑141‑3p expression. Mol Med Rep 2021; 23:259. [PMID: 33576445 PMCID: PMC7893681 DOI: 10.3892/mmr.2021.11898] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/03/2020] [Indexed: 01/21/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is caused by diabetes and can result in heart failure. Long non-coding RNAs (lncRNAs) have been demonstrated to be closely associated with DCM development. The present study aimed to investigate whether lncRNA-metastasis-associated lung adenocarcinoma transcript-1 (MALAT1) altered high glucose (HG)-induced H9C2 cardiomyocyte pyroptosis by targeting microRNA (miR)-141-3p. H9C2 cells were treated with normal glucose (NG) or HG. lncRNA-MALAT1 and miR-141-3p expression levels were determined via reverse transcription-quantitative PCR (RT-qPCR). MALAT1 and miR-141-3p knockdown and overexpression were established and confirmed via RT-qPCR. The association between MALAT1 expression and miR-141-3p expression, as well as the induction of pyroptosis and gasdermin D (GSDMD)-N expression were evaluated by performing dual luciferase reporter, TUNEL staining and immunofluorescence staining assays, respectively. Western blotting was conducted to measure the expression levels of pyroptosis-associated proteins, including apoptosis-associated speck-like protein, GSDMD-N, caspase-1, nucleotide oligomerization domain-like receptor protein 3 and GSDMD. MALAT1 mRNA expression levels were significantly increased, whereas miR-141-3p expression levels were significantly decreased in HG-treated H9C2 cells compared with the NG group. Compared with the HG group, MALAT1 overexpression significantly reduced miR-141-3p expression levels, increased the rate of TUNEL positive cells and upregulated the expression levels of pyroptosis-associated proteins. MALAT1 knockdown displayed the opposite effect on the rate of TUNEL positive cells and the expression levels of pyroptosis-associated proteins. Furthermore, the rate of TUNEL positive cells, and GSDMD-N and pyroptosis-associated protein expression levels were significantly reduced by miR-141-3p overexpression in MALAT1-overexpression H9C2 cells. The results indicated that compared with NG treatment, HG treatment increased MALAT1 expression levels and decreased miR-141-3p expression levels in H9C2 cells. Therefore, the present study suggested that lncRNA-MALAT1 targeted miR-141-3p to promote HG-induced H9C2 cardiomyocyte pyroptosis.
Collapse
Affiliation(s)
- Aishan Wu
- Department of Cardiology II, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Weili Sun
- Department of Anesthesiology, Weihai Municipal Hospital, Weihai, Shandong 264200, P.R. China
| | - Fengying Mou
- Department of Ultrasound, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
20
|
Bale S, Varga J, Bhattacharyya S. Role of RP105 and A20 in negative regulation of toll-like receptor activity in fibrosis: potential targets for therapeutic intervention. AIMS ALLERGY AND IMMUNOLOGY 2021. [DOI: 10.3934/allergy.2021009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
21
|
Guo X, Hu S, Liu J, Huang L, Zhong P, Fan Z, Ye P, Chen M. Piperine protects against pyroptosis in myocardial ischaemia/reperfusion injury by regulating the miR-383/RP105/AKT signalling pathway. J Cell Mol Med 2021; 25:244-258. [PMID: 33219747 PMCID: PMC7810957 DOI: 10.1111/jcmm.15953] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 09/12/2020] [Accepted: 09/14/2020] [Indexed: 12/18/2022] Open
Abstract
miRNA-mediated pyroptosis play crucial effects in the development of myocardial ischaemia/reperfusion (I/R) injury (MIRI). Piperine (PIP) possesses multiple pharmacological effects especially in I/R condition. This study focuses on whether PIP protects MIRI from pyroptosis via miR-383-dependent pathway. Rat MIRI model was established by 30 minutes of LAD ligation and 4 hours of reperfusion. Myocardial enzymes, histomorphology, structure and function were detected to evaluate MIRI. Recombinant adenoviral vectors for miR-383 overexpression or miR-383 silencing or RP105 knockdown were constructed, respectively. Luciferase reporter analysis was used to confirm RP105 as a target of miR-383. Pyroptosis-related markers were measured by Western blotting assay. The results showed that I/R provoked myocardial injury, as shown by the increases of LDH/CK releases, infarcted areas and apoptosis as well as worsened function and structure. Pyroptosis-related mediators including NLRP3, cleaved caspase-1, cleaved IL-1β and IL-18 were also reinforced after MIRI. However, PIP treatment greatly ameliorated MIRI in parallel with pyroptotic repression. In mechanistic studies, MIRI-caused elevation of miR-383 and decrease of RP105/PI3K/AKT pathway were reverted by PIP treatment. Luciferase reporter assay confirmed RP105 as a miR-383 target. miR-383 knockdown ameliorated but miR-383 overexpression facilitated pyroptosis and MIRI. Moreover, the anti-pyroptotic effect from miR-383 silencing was verified to be relied on the RP105/PI3K/AKT signalling pathway. Additionally, our present study further indicated the miR-383/RP105/AKT-dependent approach resulting from PIP administration against pyroptosis in MIRI. Therefore, PIP treatment attenuates MIRI and pyroptosis by regulating miR-383/RP105/AKT pathway, and it may provide a therapeutic manner for the treatment of MIRI.
Collapse
Affiliation(s)
- Xin Guo
- Department of CardiologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Shan Hu
- Heart Function DepartmentThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ji‐Jun Liu
- Department of CardiologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ling Huang
- Department of CardiologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Peng Zhong
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Zhi‐Xing Fan
- Department of CardiologyRenmin Hospital of Wuhan UniversityWuhanChina
| | - Ping Ye
- Department of CardiologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Man‐Hua Chen
- Department of CardiologyThe Central Hospital of WuhanTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
22
|
Xu Q, Xing H, Wu J, Chen W, Zhang N. miRNA-141 Induced Pyroptosis in Intervertebral Disk Degeneration by Targeting ROS Generation and Activating TXNIP/NLRP3 Signaling in Nucleus Pulpous Cells. Front Cell Dev Biol 2020; 8:871. [PMID: 32984347 PMCID: PMC7487322 DOI: 10.3389/fcell.2020.00871] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/11/2020] [Indexed: 12/12/2022] Open
Abstract
The role and mechanism of pyroptosis in intervertebral disk (IVD) degeneration are unclear. MicroRNAs (miRNAs) regulate the viability and function of nucleus pulposus cells (NPCs) in IVDs and are related to pyroptosis. We performed microarray analyses of normal and degenerated nucleus pulposus (NP) to assess the role of pyroptosis and identify key miRNAs in IVD degeneration. We also evaluated the underlying mechanism of miRNA-mediated pyroptosis in NPCs. In addition, we demonstrated the preventative effects of miRNAs on IVD degeneration in a rat model. The levels of the pyroptosis-related proteins cleaved caspase-1, N-terminal gasdermin D (GSDMD), interleukin (IL)-1β, and IL-18 in the degenerative NP were significantly higher than those in the normal NP. miRNA-141 was significantly upregulated in the degenerated NP. miR-141 mimic suppressed the matrix synthesis function of NPCs. By contrast, reactive oxygen species (ROS) generation, and the expression of TXNIP and NLRP3 were significantly downregulated by an miR-141 inhibitor. Furthermore, the miRNA-141 inhibitor prevented the degeneration of IVDs in vivo. Our findings suggest that miRNA-141 induces pyroptosis and extracellular matrix (ECM) catabolism in NPCs by increasing ROS generation and activating TXNIP/NLRP3 signaling. miRNA-141-regulated pyroptosis may be a novel therapeutic target for IVD degeneration.
Collapse
Affiliation(s)
- Qiaolong Xu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.,Department of Orthopaedics, The People's Hospital of Cixi, Cixi, China
| | - Hongyuan Xing
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jiaqi Wu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Weishan Chen
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ning Zhang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
23
|
Song J, Kim YK. Discovery and Functional Prediction of Long Non-Coding RNAs Common to Ischemic Stroke and Myocardial Infarction. J Lipid Atheroscler 2020; 9:449-459. [PMID: 33024736 PMCID: PMC7521976 DOI: 10.12997/jla.2020.9.3.449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/21/2020] [Accepted: 08/02/2020] [Indexed: 12/26/2022] Open
Abstract
Objective Ischemic stroke and myocardial infarction are 2 of the leading causes of mortality. Both conditions are caused by arterial occlusion, resulting in ischemic necrosis of the cells in the cortex and heart. Long non-coding RNAs (lncRNAs) are a group of non-coding RNAs longer than 200 nucleotides without protein-coding potential. Thousands of lncRNAs have been identified but their involvement in ischemic stroke and myocardial infarction has not been studied extensively. Therefore, this study aimed to identify the role of lncRNAs, particularly those that are commonly altered in these two ischemic injuries. Methods We combined diverse RNA sequencing data obtained from public databases and performed extensive bioinformatics analyses to determine reliable lncRNAs commonly identified from these datasets. Using sequence analysis, we also detected the lncRNAs that may act as microRNA (miRNA) regulators. Results We found several altered lncRNAs that were common in ischemic stroke and myocardial infarction models. Some of these lncRNAs, including zinc finger NFX1-type containing 1 antisense RNA 1 and small nucleolar RNA host gene 1, were previously reported to be involved in the pathogenesis of each of these models. Interestingly, several lncRNAs had binding sites for miRNAs that were previously reported to be involved in the hypoxic response, suggesting the possible role of these lncRNAs as regulators in ischemic responses. Conclusion The lncRNAs identified in this study will be useful in determining the regulatory networks in ischemic stroke and myocardial infarction and in identifying potential specific markers for each of these ischemic diseases.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Anatomy, Chonnam National University Medical School, Hwasun, Korea
| | - Young-Kook Kim
- Department of Biochemistry, Chonnam National University Medical School, Hwasun, Korea
| |
Collapse
|
24
|
Pan F, Xu X, Zhan Z, Xu Q. 6-Gingerol protects cardiomyocytes against hypoxia-induced injury by regulating the KCNQ1OT1/miR-340-5p/ PI3K/AKT pathway. Panminerva Med 2020; 63:482-490. [PMID: 32720790 DOI: 10.23736/s0031-0808.20.03956-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Hypoxia could induce cardiomyocytes injury and lead to heart disease. Studies have shown that 6-Gingerol has a protective effect on cardiomyocytes injury, but its molecular mechanism is still unclear. METHODS Cell counting kit 8 (CCK8) and flow cytometry assays were used to measure the viability and apoptosis of cardiomyocytes. Western blot (WB) analysis was performed to assess the levels of proliferation, apoptosis, and phosphatidylinositol 3- kinase/protein kinase B (PI3K/AKT) signaling pathway-related proteins. The reactive oxygen species (ROS) level, superoxide dismutase (SOD) activity and malondialdehyde (MDA) level were detected by their corresponding Assay Kits. Besides, the expression levels of potassium voltage-gated channel subfamily Q member 1 opposite strand 1 (KCNQ1OT1) and microRNA-340-5p (miR-340-5p) were determined by quantitative real-time polymerase chain reaction (qRT-PCR). Furthermore, dual-luciferase reporter and RNA immunoprecipitation (RIP) assays were used to verify the interaction between KCNQ1OT1 and miR-340-5p. RESULTS Hypoxia could inhibit the viability and enhance the apoptosis and oxidative stress of cardiomyocytes to induce cardiomyocytes injury, while 6-Gingerol could alleviate this effect. Overexpression of KCNQ1OT1 aggravated hypoxia-induced cardiomyocytes injury and reversed the protective effect of 6-Gingerol on cardiomyocytes injury. Besides, miR-340-5p could be sponged by KCNQ1OT1, and its overexpression could invert the promotion effect of KCNQ1OT1 overexpression on hypoxia-induced cardiomyocytes injury. Moreover, miR-340-5p expression was regulated by 6-Gingerol and KCNQ1OT1. In addition, hypoxia inactivated the PI3K/AKT signaling pathway, whereas 6-Gingerol and miR-340-5p could reverse this effect. CONCLUSIONS 6-Gingerol could hinder the expression of KCNQ1OT1 to protect cardiomyocytes from hypoxia-induced injury through regulation of the miR-340-5p/ PI3K/AKT pathway, providing a new mechanism of 6-Gingerol protecting cardiomyocytes from injury.
Collapse
Affiliation(s)
- Fan Pan
- Department of Internal Medicine-Cardiovascular, Putuo District People's Hospital, Shanghai, China
| | - Xiaopeng Xu
- Department of Internal Medicine-Cardiovascular, Putuo District People's Hospital, Shanghai, China
| | - Zhi Zhan
- Department of Internal Medicine-Cardiovascular, Putuo District People's Hospital, Shanghai, China
| | - Qunfeng Xu
- Department of Internal Medicine-Cardiovascular, Putuo District People's Hospital, Shanghai, China -
| |
Collapse
|
25
|
Carbonell T, Gomes AV. MicroRNAs in the regulation of cellular redox status and its implications in myocardial ischemia-reperfusion injury. Redox Biol 2020; 36:101607. [PMID: 32593128 PMCID: PMC7322687 DOI: 10.1016/j.redox.2020.101607] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/31/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022] Open
Abstract
MicroRNAs (miRNAs) are small RNAs that do not encode for proteins and play key roles in the regulation of gene expression. miRNAs are involved in a comprehensive range of biological processes such as cell cycle control, apoptosis, and several developmental and physiological processes. Oxidative stress can affect the expression levels of multiple miRNAs and, conversely, miRNAs may regulate the expression of redox sensors, alter critical components of the cellular antioxidants, interact with the proteasome, and affect DNA repair systems. The number of publications identifying redox-sensitive miRNAs has increased significantly over the last few years, and some miRNA targets such as Nrf2, SIRT1 and NF-κB have been identified. The complex interplay between miRNAs and ROS is discussed together with their role in myocardial ischemia-reperfusion injury and the potential use of circulating miRNAs as biomarkers of myocardial infarction. Detailed knowledge of redox-sensitive miRNAs is needed to be able to effectively use individual compounds or sets of miRNA-modulating compounds to improve the health-related outcomes associated with different diseases.
Collapse
Affiliation(s)
- Teresa Carbonell
- Department of Cell Biology, Physiology and Immunology, University of Barcelona, Avda Diagonal 643, 08028, Barcelona, Spain.
| | - Aldrin V Gomes
- Department of Physiology and Membrane Biology, University of California, Davis, 176 Briggs Hall, One Shields Avenue, Davis, CA, 95616, USA; Department of Physiology, Neurobiology and Behavior, University of California, Davis, 176 Briggs Hall, One Shields Avenue, Davis, CA, 95616, USA
| |
Collapse
|
26
|
Zhang X, Huang CR, Pan S, Pang Y, Chen YS, Zha GC, Guo KJ, Zheng X. Long non-coding RNA SNHG15 is a competing endogenous RNA of miR-141-3p that prevents osteoarthritis progression by upregulating BCL2L13 expression. Int Immunopharmacol 2020; 83:106425. [PMID: 32247266 DOI: 10.1016/j.intimp.2020.106425] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022]
Abstract
Increasing evidence has demonstrated that the dysregulated expression of long noncoding RNAs (lncRNAs) has important roles in the progression of osteoarthritis (OA), but the function of the lncRNA SNHG15 remains unclear. In the present study, we observed that SNHG15 was downregulated in OA cartilage tissues and IL-1β-induced chondrocytes. The lower expression of SNHG15 was negatively associated with the observed modified Mankin scale scores, extracellular matrix (ECM) degradation and chondrocyte apoptosis. Downregulated expression of SNHG15 increased chondrocyte viability and decreased chondrocyte apoptosis and ECM degradation in vitro and reduced damage to articular cartilage in vivo. Mechanistically, we demonstrated that SNHG15 overexpression promotes the expression of BCL2L13 by sponging miR-141-3p. The higher expression of miR-141-3p was negatively correlated with SNHG15 and BCL2L13 levels in OA cartilage tissues, and a positive correlation was also shown between SNHG15 and BCL2L13 levels. Furthermore, ectopic expression of miR-141-3p or knockdown of BCL2L13 expression could both reduce the effects of SNHG15 on chondrocyte proliferation, apoptosis and ECM degradation. Collectively, these findings reveal that SNHG15 inhibits OA progression by acting as an miR-141-3p sponge to promote BCL2L13 expression, suggesting that knockdown of SNHG15 expression in chondrocytes can be a potential therapeutic strategy to ameliorate OA progression.
Collapse
Affiliation(s)
- Xing Zhang
- Department of Orthopaedics, Jintan Hospital Affiliated to Jiangsu University, No. 16 South Gate Street, Jintan, Jiangsu 213200, China
| | - Chao-Ran Huang
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road West, Xuzhou, Jiangsu 221006, China
| | - Sheng Pan
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road West, Xuzhou, Jiangsu 221006, China
| | - Yong Pang
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road West, Xuzhou, Jiangsu 221006, China
| | - Ye-Shuai Chen
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road West, Xuzhou, Jiangsu 221006, China
| | - Guo-Chun Zha
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road West, Xuzhou, Jiangsu 221006, China
| | - Kai-Jin Guo
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road West, Xuzhou, Jiangsu 221006, China
| | - Xin Zheng
- Department of Orthopaedics, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road West, Xuzhou, Jiangsu 221006, China.
| |
Collapse
|
27
|
Wei L, Chen Q, Zheng Y, Nan L, Liao N, Mo S. Potential Role of Integrin α₅β₁/Focal Adhesion Kinase (FAK) and Actin Cytoskeleton in the Mechanotransduction and Response of Human Gingival Fibroblasts Cultured on a 3-Dimension Lactide-Co-Glycolide (3D PLGA) Scaffold. Med Sci Monit 2020; 26:e921626. [PMID: 32034900 PMCID: PMC7027369 DOI: 10.12659/msm.921626] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The stability of orthodontic treatment is thought to be significantly affected by the compression and retraction of gingival tissues, but the underlying molecular mechanism is not fully elucidated. The objectives of our study were to explore the effects of mechanical force on the ECM-integrin-cytoskeleton linkage response in human gingival fibroblasts (HGFs) cultured on 3-dimension (3D) lactide-co-glycolide (PLGA) biological scaffold and to further study the mechanotransduction pathways that could be involved. MATERIAL AND METHODS A compressive force of 25 g/m² was applied to the HGFs-PLGA 3D co-cultured model. Rhodamine-phalloidin staining was used to evaluate the filamentous actin (F-actin) cytoskeleton. The expression level of type I collagen (COL-1) and the activation of the integrin alpha₅ß₁/focal adhesion kinase (FAK) signaling pathway were determined by using real-time PCR and Western blotting analysis. The impacts of the applied force on the expression levels of FAK, phosphorylated focal adhesion kinase (p-FAK), and COL-1 were also measured in cells treated with integrin alpha₅ß₁ inhibitor (Ac-PHSCN-NH 2, ATN-161). RESULTS Mechanical force increased the expression of integrin alpha₅ß₁, FAK (p-FAK), and COL-1 in HGFs, and induced the formation of stress fibers. Blocking integrin alpha₅ß₁ reduced the expression of FAK (p-FAK), while the expression of COL-1 was not fully inhibited. CONCLUSIONS The integrin alpha₅ß₁/FAK signaling pathway and actin cytoskeleton appear to be involved in the mechanotransduction of HGFs. There could be other mechanisms involved in the promotion effect of mechanical force on collagen synthesis in addition to the integrin alpha₅ß₁ pathway.
Collapse
Affiliation(s)
- Liying Wei
- Department of Stomatology, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Clinical Research Center for Craniofacial Deformity, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Qun Chen
- Department of Stomatology, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Key Laboratory of Oral and Maxillofacial Surgery Disease Treatment, Guangxi Medical University, Nanning, Guangxi, China (mainland).,Clinical Research Center for Craniofacial Deformity, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Yi Zheng
- Department of Stomatology, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Lan Nan
- Department of Stomatology, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Ni Liao
- Department of Stomatology, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Shuixue Mo
- Department of Stomatology, Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|