1
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
2
|
Alvia M, Aytan N, Spencer KR, Foster ZW, Rauf NA, Guilderson L, Robey I, Averill JG, Walker SE, Alvarez VE, Huber BR, Mathais R, Cormier KA, Nicks R, Pothast M, Labadorf A, Agus F, Alosco ML, Mez J, Kowall NW, McKee AC, Brady CB, Stein TD. MicroRNA Alterations in Chronic Traumatic Encephalopathy and Amyotrophic Lateral Sclerosis. Front Neurosci 2022; 16:855096. [PMID: 35663558 PMCID: PMC9160996 DOI: 10.3389/fnins.2022.855096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/09/2022] [Indexed: 11/13/2022] Open
Abstract
Repetitive head impacts (RHI) and traumatic brain injuries are risk factors for the neurodegenerative diseases chronic traumatic encephalopathy (CTE) and amyotrophic lateral sclerosis (ALS). ALS and CTE are distinct disorders, yet in some instances, share pathology, affect similar brain regions, and occur together. The pathways involved and biomarkers for diagnosis of both diseases are largely unknown. MicroRNAs (miRNAs) involved in gene regulation may be altered in neurodegeneration and be useful as stable biomarkers. Thus, we set out to determine associations between miRNA levels and disease state within the prefrontal cortex in a group of brain donors with CTE, ALS, CTE + ALS and controls. Of 47 miRNAs previously implicated in neurological disease and tested here, 28 (60%) were significantly different between pathology groups. Of these, 21 (75%) were upregulated in both ALS and CTE, including miRNAs involved in inflammatory, apoptotic, and cell growth/differentiation pathways. The most significant change occurred in miR-10b, which was significantly increased in ALS, but not CTE or CTE + ALS. Overall, we found patterns of miRNA expression that are common and unique to CTE and ALS and that suggest shared and distinct mechanisms of pathogenesis.
Collapse
Affiliation(s)
- Marcela Alvia
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Nurgul Aytan
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | | | | | | | | | - Ian Robey
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - James G. Averill
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - Sean E. Walker
- Southern Arizona VA Healthcare System, Tucson, AZ, United States
| | - Victor E. Alvarez
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Bertrand R. Huber
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Rebecca Mathais
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Kerry A. Cormier
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Raymond Nicks
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Morgan Pothast
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
| | - Adam Labadorf
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Filisia Agus
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Michael L. Alosco
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Jesse Mez
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
| | - Neil W. Kowall
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
| | - Ann C. McKee
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| | - Christopher B. Brady
- Department of Neurology, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
| | - Thor D. Stein
- Boston University Alzheimer’s Disease Research Center, Boston University CTE Center, Boston University School of Medicine, Boston, MA, United States
- VA Boston Healthcare System, Boston, MA, United States
- Department of Veterans Affairs Medical Center, Bedford, MA, United States
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, MA, United States
| |
Collapse
|
3
|
Li J, Wang N, Nie H, Wang S, Jiang T, Ma X, Liu W, Tian K. Long Non-coding RNA RMST Worsens Ischemic Stroke via MicroRNA-221-3p/PIK3R1/TGF-β Signaling Pathway. Mol Neurobiol 2022; 59:2808-2821. [PMID: 35217983 DOI: 10.1007/s12035-021-02632-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 11/01/2021] [Indexed: 11/26/2022]
Abstract
Much efforts have been made to probe the mechanism underlying ischemic stroke (IS). This study was proposed to uncover the role of long non-coding RNA rhabdomyosarcoma 2 related transcript (RMST) in IS through microRNA-221-3p (miR-221-3p)/phosphoinositide-3-kinase regulatory subunit 1 (PIK3R1)/transforming growth factor-β (TGF-β) axis. Neurological behavioral function, pathological changes in brain tissue, oxidative stress, and inflammation responses in middle cerebral artery occlusion (MCAO) mice were tested. RMST, miR-221-3p, PIK3R1, and TGF-β signaling-related protein expression in brain tissues of MCAO mice were detected. RMST and PIK3R1 were elevated, miR-221-3p was downregulated, and TGF-β pathway was activated in mice after MCAO. Restored miR-221-3p or depleted RMST improved neurological behavioral functions, relieved pathological injury in brain tissue, and repressed oxidative stress and inflammation in mice after MCAO. Depleted PIK3R1 or restored miR-221-3p offsets the negative effects of overexpressed RMST on mice with MCAO. The present work highlights that RMST augments IS through reducing miR-221-3p-mediated regulation of PIK3R1 and activating TGF-β pathway.
Collapse
Affiliation(s)
- Jie Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Ning Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Huan Nie
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Shan Wang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Tongtong Jiang
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Xuehan Ma
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China
| | - Wenjuan Liu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China.
| | - Kuo Tian
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang Province, Harbin, 150081, China.
| |
Collapse
|
4
|
Florijn BW, Bijkerk R, Kruyt ND, van Zonneveld AJ, Wermer MJH. Sex-Specific MicroRNAs in Neurovascular Units in Ischemic Stroke. Int J Mol Sci 2021; 22:11888. [PMID: 34769320 PMCID: PMC8585074 DOI: 10.3390/ijms222111888] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/25/2021] [Accepted: 10/27/2021] [Indexed: 12/24/2022] Open
Abstract
Accumulating evidence pinpoints sex differences in stroke incidence, etiology and outcome. Therefore, more understanding of the sex-specific mechanisms that lead to ischemic stroke and aggravation of secondary damage after stroke is needed. Our current mechanistic understanding of cerebral ischemia states that endothelial quiescence in neurovascular units (NVUs) is a major physiological parameter affecting the cellular response to neuron, astrocyte and vascular smooth muscle cell (VSMC) injury. Although a hallmark of the response to injury in these cells is transcriptional activation, noncoding RNAs such as microRNAs exhibit cell-type and context dependent regulation of gene expression at the post-transcriptional level. This review assesses whether sex-specific microRNA expression (either derived from X-chromosome loci following incomplete X-chromosome inactivation or regulated by estrogen in their biogenesis) in these cells controls NVU quiescence, and as such, could differentiate stroke pathophysiology in women compared to men. Their adverse expression was found to decrease tight junction affinity in endothelial cells and activate VSMC proliferation, while their regulation of paracrine astrocyte signaling was shown to neutralize sex-specific apoptotic pathways in neurons. As such, these microRNAs have cell type-specific functions in astrocytes and vascular cells which act on one another, thereby affecting the cell viability of neurons. Furthermore, these microRNAs display actual and potential clinical implications as diagnostic and prognostic biomarkers in ischemic stroke and in predicting therapeutic response to antiplatelet therapy. In conclusion, this review improves the current mechanistic understanding of the molecular mechanisms leading to ischemic stroke in women and highlights the clinical promise of sex-specific microRNAs as novel diagnostic biomarkers for (silent) ischemic stroke.
Collapse
Affiliation(s)
- Barend W. Florijn
- Department of Neurology, Leiden University Medical Center, 2333 ZR Leiden, The Netherlands; (N.D.K.); (M.J.H.W.)
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.B.); (A.J.v.Z.)
| | - Roel Bijkerk
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.B.); (A.J.v.Z.)
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Nyika D. Kruyt
- Department of Neurology, Leiden University Medical Center, 2333 ZR Leiden, The Netherlands; (N.D.K.); (M.J.H.W.)
| | - Anton Jan van Zonneveld
- Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands; (R.B.); (A.J.v.Z.)
- Department of Internal Medicine (Nephrology), Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Marieke J. H. Wermer
- Department of Neurology, Leiden University Medical Center, 2333 ZR Leiden, The Netherlands; (N.D.K.); (M.J.H.W.)
| |
Collapse
|
5
|
Yao J, Du Y, Liu J, Gareev I, Yang G, Kang X, Wang X, Beylerli O, Chen X. Hypoxia related long non-coding RNAs in ischemic stroke. Noncoding RNA Res 2021; 6:153-158. [PMID: 34703955 PMCID: PMC8511691 DOI: 10.1016/j.ncrna.2021.10.001] [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: 09/09/2021] [Revised: 10/02/2021] [Accepted: 10/02/2021] [Indexed: 12/22/2022] Open
Abstract
With high rates of mortality and disability, stroke has caused huge social burden, and 85% of which is ischemic stroke. In recent years, it is a progressive discovery of long non-coding RNA (lncRNA) playing an important regulatory role throughout ischemic stroke. Hypoxia, generated from reduction or interruption of cerebral blood flow, leads to changes in lncRNA expression, which then influence disease progression. Therefore, we reviewed studies on expression of hypoxia-related lncRNAs and relevant molecular mechanism in ischemic stroke. Considering that hypoxia-inducible factor (HIF) is a crucial regulator in hypoxic progress, we mainly focus on the HIF-related lncRNA which regulates the expression of HIF or is regulated by HIF, further reveal their pathogenesis and adaption after brain ischemia and hypoxia, so as to find effective biomarker and therapeutic targets.
Collapse
Affiliation(s)
- Jiawei Yao
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Yiming Du
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Junsi Liu
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Ilgiz Gareev
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Guang Yang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Xiaohui Kang
- Department of Pharmacy, Rizhao People's Hospital, Rizhao, 276826, Shandong Province, China
| | - Xiaoxiong Wang
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| | - Ozal Beylerli
- Bashkir State Medical University, Ufa, Republic of Bashkortostan, 450008, Russia
| | - Xin Chen
- Department of Neurosurgery, First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang Province, China.,Institute of Neuroscience, Sino-Russian Medical Research Center, Harbin Medical University, Harbin, 150001, Heilongjiang Province, China
| |
Collapse
|
6
|
Ma W, Zhang X, Liu Y. miR-124 promotes apoptosis and inhibits the proliferation of vessel endothelial cells through P38/MAPK and PI3K/AKT pathways, making it a potential mechanism of vessel endothelial injury in acute myocardial infarction. Exp Ther Med 2021; 22:1383. [PMID: 34650631 PMCID: PMC8506947 DOI: 10.3892/etm.2021.10819] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 08/11/2021] [Indexed: 11/26/2022] Open
Abstract
Due to its rapid onset and high rates of fatality, acute myocardial infarction (AMI) has long been one of the most fatal diseases among all types of heart diseases. Therefore, intensive research efforts have been focused on understanding AMI's potential pathogenesis to seek effective treatment options. In the present study, 20 peripheral blood samples were collected from patients with AMI, after which reverse transcription-quantitative PCR analysis revealed that microRNA (miR)-124 levels in the peripheral blood of patients with AMI was significantly elevated compared with that in the control group. In vitro, a model using pcDNA3.1-miR-124 transfected human umbilical vein endothelial cells (HUVECs) indicated that overexpression of miR-124 could significantly promote the apoptosis and suppress the proliferation of HUVECs using flow cytometry, TUNEL assay and Cell Counting Kit-8 assays. Based on the present findings, RNA samples of HUVECs overexpressing miR-124 was extracted and sequenced to explore the gene expression profile after miR-124 overexpression. Gene Set Enrichment Analysis (GSEA) analysis revealed that the downregulated genes were mainly enriched in signaling pathways, such as PI3K-AKT, whilst the upregulated genes were mainly enriched in metabolism-related signaling pathways, such as the metabolism of xenobiotics by cytochrome P450 pathway. Additionally, Rideogram software was used to determine the chromosomal localization of the differentially expressed genes. The results demonstrated that they were distributed on all chromosomes except for chromosome Y. In addition, characteristic profiles of the differentially expressed genes caused by miR-124 overexpression were analyzed using Connectivity Map. In total, two medicines, anisomycin and sanguinarine, which function as p38/MAPK signaling agonists that can inhibit angiogenesis, presented with the highest enrichment scores. Together with the GSEA results, which indicated that the differentially expressed genes were mainly enriched in the angiogenesis-inhibiting PI3K/AKT signaling pathway, the present study reported that high expression of miR-124 was negatively associated with patients with AMI, promoting the apoptosis and suppressing the proliferation of vessel endothelial cells.
Collapse
Affiliation(s)
- Weimin Ma
- Department of Critical Care Medicine, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| | - Xin Zhang
- Department of Pediatrics, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| | - Yang Liu
- Department of Critical Care Medicine, Weihai Central Hospital, Weihai, Shandong 264200, P.R. China
| |
Collapse
|
7
|
Zhang Y, Long J, Ren J, Huang X, Zhong P, Wang B. Potential Molecular Biomarkers of Vestibular Schwannoma Growth: Progress and Prospects. Front Oncol 2021; 11:731441. [PMID: 34646772 PMCID: PMC8503266 DOI: 10.3389/fonc.2021.731441] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Accepted: 09/06/2021] [Indexed: 12/25/2022] Open
Abstract
Vestibular schwannomas (VSs, also known as acoustic neuromas) are relatively rare benign brain tumors stem from the Schwann cells of the eighth cranial nerve. Tumor growth is the paramount factor for neurosurgeons to decide whether to choose aggressive treatment approach or careful follow-up with regular magnetic resonance imaging (MRI), as surgery and radiation can introduce significant trauma and affect neurological function, while tumor enlargement during long-term follow-up will compress the adjacent nerves and tissues, causing progressive hearing loss, tinnitus and vertigo. Recently, with the deepening research of VS biology, some proteins that regulate merlin conformation changes, inflammatory cytokines, miRNAs, tissue proteins and cerebrospinal fluid (CSF) components have been proposed to be closely related to tumor volume increase. In this review, we discuss advances in the study of biomarkers that associated with VS growth, providing a reference for exploring the growth course of VS and determining the optimal treatment strategy for each patient.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Jianfei Long
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Junwei Ren
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiang Huang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ping Zhong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Bin Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
8
|
Guan X, Zhang Y, Gareev I, Beylerli O, Li X, Lu G, Lv L, Hai X. MiR-499a prevents astrocytes mediated inflammation in ischemic stroke by targeting PTEN. Noncoding RNA Res 2021; 6:146-152. [PMID: 34632168 PMCID: PMC8488463 DOI: 10.1016/j.ncrna.2021.09.002] [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: 09/09/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION Ischemic stroke (IS) is a common and severe neurological disorder and is associated with high rates of mortality and morbidity. Inflammatory reaction in astrocytes is one of the important pathological factors of stroke. Improved understanding of the underlying molecular mechanisms should aid better treatment of the disease. This study aimed to test our hypothesis that a miR-499a played an important role in the inflammatory response in astrocytes induced by IS targeting phosphatase and tensin homologue deleted on chromosome 10 (PTEN). METHODS This study was comprised of two models: oxygen-glucose deprivation (OGD) and reoxygenation model. Quantitative real-time PCR (qRT-PCR) and Western blot were used to examine gene expression levels, and MTT assay analysis were used to examine cell states. The relationships between miR-499a and PTEN were confirmed by luciferase reporter assay. RESULTS MiR-499a was robustly downregulated with OGD induced injury in astrocytes. Forced transient expression of miR-499a in OGD astrocytes nearly completely reversed the inflammatory response. Knockdown of miR-499a by its specific inhibitor in healthy astrocytes induced the inflammatory response resembling those produced by OGD. On the other hand, PTEN was markedly upregulated in OGD astrocytes, which was reciprocal to the expression of miR-499a. PTEN was experimentally validated as a direct target gene for miR-499a. Overexpression of PTEN was able to induce an inflammatory response of astrocytes. Moreover, PTEN siRNA counteracted the inflammatory response induced by OGD. CONCLUSIONS Taken together, our findings indicate miR-499a as an important factor to prevent inflammatory response and suggest miR-499a as a new molecule for the treatment of IS. The present study also demonstrated the relationship between miR-499a and PTEN, with PTEN as a downstream signaling mediator of miR-499a in the inflammatory response of astrocytes induced by IS.
Collapse
Affiliation(s)
- Xiaoxiang Guan
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Yiwei Zhang
- Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, 150001, PR China
| | - Ilgiz Gareev
- Bashkir State Medical University, Ufa, 450008, Russia
| | - Ozal Beylerli
- Bashkir State Medical University, Ufa, 450008, Russia
| | - Xinyuan Li
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Guitian Lu
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Lin Lv
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Xin Hai
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| |
Collapse
|
9
|
Endothelial Cells as Tools to Model Tissue Microenvironment in Hypoxia-Dependent Pathologies. Int J Mol Sci 2021; 22:ijms22020520. [PMID: 33430201 PMCID: PMC7825710 DOI: 10.3390/ijms22020520] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/27/2020] [Accepted: 01/05/2021] [Indexed: 12/11/2022] Open
Abstract
Endothelial cells (ECs) lining the blood vessels are important players in many biological phenomena but are crucial in hypoxia-dependent diseases where their deregulation contributes to pathology. On the other hand, processes mediated by ECs, such as angiogenesis, vessel permeability, interactions with cells and factors circulating in the blood, maintain homeostasis of the organism. Understanding the diversity and heterogeneity of ECs in different tissues and during various biological processes is crucial in biomedical research to properly develop our knowledge on many diseases, including cancer. Here, we review the most important aspects related to ECs’ heterogeneity and list the available in vitro tools to study different angiogenesis-related pathologies. We focus on the relationship between functions of ECs and their organo-specificity but also point to how the microenvironment, mainly hypoxia, shapes their activity. We believe that taking into account the specific features of ECs that are relevant to the object of the study (organ or disease state), especially in a simplified in vitro setting, is important to truly depict the biology of endothelium and its consequences. This is possible in many instances with the use of proper in vitro tools as alternative methods to animal testing.
Collapse
|
10
|
Zhou M, Li C. Clinical Value and Potential Target of miR-27a-3p in Pulpitis. Neuroimmunomodulation 2021; 28:158-165. [PMID: 34237753 DOI: 10.1159/000516136] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 03/23/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION This study investigated the clinical values of miR-27a-3p for pulpitis patients, and its association with TLR4. METHODS Sixty-six patients with pulpitis and 34 cases without pulpitis were recruited; the pulp tissue and serum samples were collected from each participant. Real-time polymerase chain reaction was used for measurement of gene expression levels. The diagnosis values were assessed by the receiver operating characteristic curve. The target gene of miR-27a-3p was confirmed by the luciferase reporter assay. RESULTS MiR-27a-3p was downregulated in both serum and pulp tissue of pulpitis patients. MiR-27a-3p could distinguish pulpitis patients from healthy controls and might be a predictor for the development of irreversible pulpitis. A high level of TLR4 was also detected in both peripheral blood monocytes and pulp tissues from pulpitis patients and showed a negative association with the miR-27a-3p level. TLR4 was a direct target gene of miR-27a-3p. DISCUSSION/CONCLUSION MiR-27a-3p might be a promising biomarker for the diagnosis of pulpitis and predict the development of irreversible pulpitis. MiR-27a-3p might be involved in the pathogenesis of pulpitis via targeting TLR4.
Collapse
Affiliation(s)
- Ming Zhou
- Department of Endodontics, Stomatology Hospital of Wuhan University Qingshan Branch, Wuhan, China
| | - Chaohong Li
- Department of Prosthodontics, Stomatology Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
11
|
Yu M, Liu W, Li J, Lu J, Lu H, Jia W, Liu F. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther 2020; 11:350. [PMID: 32787917 PMCID: PMC7425015 DOI: 10.1186/s13287-020-01824-2] [Citation(s) in RCA: 239] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/20/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Background Mesenchymal stem cell (MSC)-derived exosomes emerge as promising candidates for treating delayed wound healing in diabetes due to the promotion of angiogenesis. Preconditioned MSC with chemical or biological factors could possibly enhance the biological activities of MSC-derived exosomes. The purpose of this research focused on whether exosomes derived from the bone marrow MSC (BMSC) pretreated with atorvastatin (ATV), could exhibit better pro-angiogenic ability in diabetic wound healing or not and its underlying molecular mechanism. Methods We isolated exosomes from non-pretreated BMSC (Exos) and ATV pretreated BMSC (ATV-Exos) and evaluated their characterization by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA) and Western blotting. In vivo, we made full-thickness skin defects in streptozotocin (STZ)-induced diabetic rats and the defects received multiple-point injection with PBS, Exos, or ATV-Exos. Two weeks later, histological analysis was conducted to evaluate the impact of different treatments on wound healing and the neovascularization was measured by micro-CT. In vitro, cell proliferation, migration, tube formation, and vascular endothelial growth factor (VEGF) secretion were measured in human umbilical vein endothelial cells (HUVEC). The role of miRNAs and AKT/eNOS signaling pathway in the promoted angiogenesis of ATV-Exos were assessed with their inhibitors. Results No significant difference in morphology, structure, and concentration was observed between ATV-Exos and Exos. In STZ-induced diabetic rats, ATV-Exos exhibited excellent abilities in facilitating the wound regeneration by promoting the formation of blood vessels compared with Exos without influencing liver and kidney function. Meanwhile, ATV-Exos promoted the proliferation, migration, tube formation, and VEGF level of endothelial cells in vitro. And AKT/eNOS pathway was activated by ATV-Exos and the pro-angiogenic effects of ATV-Exo were attenuated after the pathway being blocked. MiR-221-3p was upregulated by ATV-Exos stimulation, and miR-221-3p inhibitor suppressed the pro-angiogenesis effect of ATV-Exos. Conclusions Exosomes originated from ATV-pretreated MSCs might serve as a potential strategy for the treatment of diabetic skin defects through enhancing the biological function of endothelial cells via AKT/eNOS pathway by upregulating the miR-221-3p.
Collapse
Affiliation(s)
- Muyu Yu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Wei Liu
- Spine Center, Department of Orthopaedics, Shanghai Changzheng Hospital, Second Military Medical University, Shanghai, 200003, China
| | - Junxian Li
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Junxi Lu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Huijuan Lu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Fang Liu
- Department of Endocrinology and Metabolism, Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
12
|
Gao XZ, Ma RH, Zhang ZX. miR-339 Promotes Hypoxia-Induced Neuronal Apoptosis and Impairs Cell Viability by Targeting FGF9/CACNG2 and Mediating MAPK Pathway in Ischemic Stroke. Front Neurol 2020; 11:436. [PMID: 32587563 PMCID: PMC7297914 DOI: 10.3389/fneur.2020.00436] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/23/2020] [Indexed: 12/20/2022] Open
Abstract
Ischemic stroke (IS) is a common cerebrovascular disease characterized by insufficient blood blow to the brain and the second leading cause of death as well as disability worldwide. Recent literatures have indicated that abnormal expression of miR-339 is closely related to IS. In this study, we attempted to assess the biological function of miR-339 and its underlying mechanism in IS. By accessing the GEO repository, the expression of miR-339, FGF9, and CACNG2 in middle cerebral artery occlusion (MCAO) and non-MCAO was evaluated. PC12 cells after oxygen-glucose deprivation/reoxygenation (OGD/R) treatment were prepared to mimic in vitro the IS model. The levels of miR-339, FGF9, CACNG2, and MAPK-related markers were quantitatively measured by qRT-PCR and Western blot. CCK-8 and flow cytometry analyses were performed to examine cell viability and apoptosis, respectively. IS-related potential pathways were identified using KEGG enrichment analysis and GO annotations. Bioinformatics analysis and dual-luciferase reporter assay were used to predict and verify the possible target of miR-339. Our results showed that miR-339 expression was significantly increased in MCAO and OGD/R-treated PC12 cells. Overexpression of miR-339 inhibited cell viability of PC12 cells subjected to OGD/R treatment. FGF9 and CACMG2 are direct targets of miR-339 and can reverse the aggressive effect of miR-339 on the proliferation and apoptosis of OGD/R-treated PC12 cells. Moreover, miR-339 mediated the activation of the MAPK pathway, which was inhibited by the FGF9/CACNG2 axis in PC12 cells treated by OGD/R stimulation. In summary, these findings suggested that miR-339 might act as a disruptive molecule to accelerate the IS progression via targeting the FGF9/CACNG2 axis and mediating the MAPK pathway.
Collapse
Affiliation(s)
- Xiao-Zeng Gao
- Department of Anesthesiology, North China University of Science and Technology, Tangshan, China
| | - Ru-Hua Ma
- Emergency Department, Rizhao Hospital of Traditional Chinese Medicine, Rizhao, China
| | - Zhao-Xia Zhang
- Department of Geriatrics, Shanxian Central Hospital, Heze, China
| |
Collapse
|