1
|
Sharon Y, Ben-David G, Nisgav Y, Amarilyo G, Shapira G, Israel-Elgali I, Pillar S, Pillar N, Shomron N, Kramer M. MicroRNAs as Biomarkers for Uveitis in Juvenile Idiopathic Arthritis. Ocul Immunol Inflamm 2025; 33:589-595. [PMID: 39561030 DOI: 10.1080/09273948.2024.2428417] [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: 07/26/2024] [Revised: 09/22/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024]
Abstract
PURPOSE Uveitis associated with juvenile idiopathic arthritis (JIA-U) is a clinically silent vision-impairing disease. Early detection and aggressive treatment are crucial for optimal visual outcome. Alterations in levels of microRNAs (miRNAs) are characteristic of autoimmune diseases. The present clinical study sought to explore the expression of miRNAs in JIA-U and their potential role as a predictive biomarker. METHODS MiRNA expression profiling was performed on peripheral blood mononuclear cells derived from pediatric patients with JIA, JIA-U, or other types of uveitis using the high-throughput small-RNA sequencing (on Next Generation Sequencing (NGS)). Patient- and disease-related data were retrieved from the medical files. Main outcome measure was the differential expression of miRNAs among the groups. RESULTS The cohort included 35 patients; 20 children with JIA-U (8 with active disease), 10 with JIA without ocular involvement, and 5 with other types of uveitis (4 with active disease). Mean age was 8.6 years; 83% were female. Nineteen patients (54%) received immunomodulatory treatment. The expression of miR-4485-3p was significantly increased in patients with JIA-U compared to patients with JIA alone (p < 0.05), with no difference between patients with active or inactive uveitis. The expression in patients with uveitis of other etiologies was similar to the expression in JIA-U patients. CONCLUSIONS This study demonstrates a differential expression profile of a specific miRNA in JIA patients with and without uveitis. If verified in larger studies, the findings may assist to identify JIA patients at risk to develop uveitis and to improve early detection of disease activity.
Collapse
Affiliation(s)
- Yael Sharon
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gil Ben-David
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yael Nisgav
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Laboratory of Eye Research, Felsenstein Medical Research Center, Petach Tikva, Israel
| | - Gil Amarilyo
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Department of Pediatric Rheumatology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Guy Shapira
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ifat Israel-Elgali
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shani Pillar
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nir Pillar
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Noam Shomron
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Michal Kramer
- Department of Ophthalmology, Rabin Medical Center, Beilinson Hospital, Petach Tikva, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
2
|
Gambari R, Papi C, Gasparello J, Agostinelli E, Finotti A. Preliminary results and a theoretical perspective of co‑treatment using a miR‑93‑5p mimic and aged garlic extract to inhibit the expression of the pro‑inflammatory interleukin‑8 gene. Exp Ther Med 2025; 29:85. [PMID: 40084194 PMCID: PMC11904878 DOI: 10.3892/etm.2025.12835] [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: 10/08/2024] [Accepted: 01/07/2025] [Indexed: 03/16/2025] Open
Abstract
The coronavirus disease-19 (COVID-19) pandemic has been a very significant health issue in the period between 2020 and 2023, forcing research to characterize severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) sequences and to develop novel therapeutic approaches. Interleukin-6 (IL-6) and IL-8 are considered significant therapeutic targets for COVID-19 and emerging evidence has suggested that microRNAs (miRNAs/miRs) serve a key role in regulating these genes. MiRNAs are short, 19-25 nucleotides in length, non-coding RNAs that regulate gene expression at the post-transcriptional level through the sequence-selective recognition of the 3'-untranslated region (3'-UTR) of the regulated mRNAs, eventually repressing translation, commonly, via mRNA degradation. For example, among several miRNAs involved in the regulation of the COVID-19 'cytokine storm', miR-93-5p can inhibit IL-8 gene expression by directly targeting the 3'-UTR of IL-8 mRNA. In addition, miR-93-5p can regulate Toll-like receptor-4 (TLR4) and interleukin-1 receptor-associated kinase 4 (IRAK4) expression, thus affecting the nuclear factor-κB (NF-κB) pathway and the expression of NF-κB-regulated genes, such as IL-6, IL-1β and other hyper-expressed genes during the COVID-19 'cytokine storm'. In the present study, the results provided preliminary evidence suggesting that the miR-93-5p-based miRNA therapeutics could be combined with the anti-inflammatory aged garlic extract (AGE) to more effectively inhibit IL-8 gene expression. The human bronchial epithelial IB3-1 cell line was employed as experimental model system. IB3-1 cells were stimulated with the BNT162b2 COVID-19 vaccine and transfected with pre-miR-93-5p in the absence or in the presence of AGE, to verify the inhibitory effects on the BNT162b2-induced expression of the IL-8 gene. The accumulation of IL-8 mRNA was assessed by RT-qPCR; the release of IL-8 protein was determined by Bio-Plex assay. In addition, the possible applications of TLR4/NF-κB inhibitory agents (such as miR-93-5p and AGE) for treating human pathologies at a hyperinflammatory state, such as COVID-19, cystic fibrosis and other respiratory diseases, were summarized.
Collapse
Affiliation(s)
- Roberto Gambari
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Chiara Papi
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Jessica Gasparello
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| | - Enzo Agostinelli
- Department of Sensory Organs, Sapienza University of Rome, Policlinico Umberto I, I-00161 Rome, Italy
- International Polyamines Foundation ‘ETS-ONLUS’, I-00159 Rome, Italy
| | - Alessia Finotti
- Department of Life Sciences and Biotechnology, University of Ferrara, I-44121 Ferrara, Italy
| |
Collapse
|
3
|
Bargues-Carot A, Prado-Rico J, Kawasawa YI, Cai J, Yanosky JD, Zenitsky G, Jin H, Lewis M, Ma P, Anantharam V, Kanthasamy A, Rico ALG, Hall MA, Mailman RB, Kanthasamy AG, Huang X. MicroRNA Expression in Asymptomatic Welders: Implications for Biomarker Discovery for Environmentally-Linked Neurodegenerative Disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.10.25322027. [PMID: 39990579 PMCID: PMC11844587 DOI: 10.1101/2025.02.10.25322027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Chronic occupational exposure to metals in welding fumes has been implicated in the etiology of neurodegenerative diseases (NDDs), including Parkinson's disease (PD) and Alzheimer's disease (AD). Changes in microRNA (miRNA) expression have been associated with various neurodegenerative conditions. Circulating miRNAs, in particular, have emerged as promising, minimally invasive biomarkers for diagnosing and monitoring disease progression. This study was designed to characterize the expression of miRNAs in neuronally-enriched serum extracellular vesicles (EVs) among welders and non-welders to explore their potential link to metal concentrations and welding exposure measures and their potential as early diagnostic biomarkers for neurodegeneration. Serum samples from 39 welders and 27 healthy individuals were collected, and EV-enclosed miRNAs were extracted and analyzed. Also, whole blood metal concentrations and welding exposure measurements were obtained. Fifty miRNAs were found to be dysregulated in welders vs. non-welders, of which three (miR-16-5p, miR-93-5p, miR-486-5p) showing reduced expression and two (miR-4281 and miR-4417) exhibiting positive correlations with blood metal concentrations as well as with long- and short-term welding exposure measures. The dysregulation of these miRNAs suggests that exposure to metals could disrupt important biological processes, possibly contributing to an elevated risk of NDDs. These findings highlight the need for further research to validate the causal relationship between exposure to metals in welding fumes, the dysregulation of circulating miRNAs, and their role in neurodegenerative disease development, with implications for miRNA-based biomarkers in early disease detection and prevention.
Collapse
Affiliation(s)
- Alejandra Bargues-Carot
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Janina Prado-Rico
- Departments of Neurology and of Neuroscience & Experimental Therapeutics, Translational Brain Research Center, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Yuka Imamura Kawasawa
- Departments of Neurology and of Neuroscience & Experimental Therapeutics, Translational Brain Research Center, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Jiazhang Cai
- Department of Public Health Sciences, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA. Department of Statistics, University of Georgia, Athens, GA, USA
| | - Jeff D. Yanosky
- Department of Public Health Sciences, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA. Department of Statistics, University of Georgia, Athens, GA, USA
| | - Gary Zenitsky
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Huajun Jin
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Mechelle Lewis
- Departments of Neurology and of Neuroscience & Experimental Therapeutics, Translational Brain Research Center, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Ping Ma
- Department of Public Health Sciences, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA. Department of Statistics, University of Georgia, Athens, GA, USA
| | - Vellareddy Anantharam
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Arthi Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | | | - Molly A. Hall
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard B. Mailman
- Departments of Neurology and of Neuroscience & Experimental Therapeutics, Translational Brain Research Center, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Anumantha G. Kanthasamy
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, University of Georgia, Athens, GA, USA
| | - Xuemei Huang
- Departments of Neurology and of Neuroscience & Experimental Therapeutics, Translational Brain Research Center, Pennsylvania State University College of Medicine and Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
4
|
Saadat M, Dahmardeh N, Sheikhbahaei F, Mokhtari T. Therapeutic potential of thymoquinone and its nanoformulations in neuropsychological disorders: a comprehensive review on molecular mechanisms in preclinical studies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:3541-3564. [PMID: 38010395 DOI: 10.1007/s00210-023-02832-8] [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: 12/12/2022] [Accepted: 10/30/2023] [Indexed: 11/29/2023]
Abstract
Thymoquinone (THQ) and its nanoformulation (NFs) have emerged as promising candidates for the treatment of neurological diseases due to their diverse pharmacological properties, which include anti-inflammatory, antioxidant, and neuroprotective effects. In this study, we conducted an extensive search across reputable scientific websites such as PubMed, ScienceDirect, Scopus, and Google Scholar to gather relevant information. The antioxidant and anti-inflammatory properties of THQ have been observed to enhance the survival of neurons in affected areas of the brain, leading to significant improvements in behavioral and motor dysfunctions. Moreover, THQ and its NFs have demonstrated the capacity to restore antioxidant enzymes and mitigate oxidative stress. The primary mechanism underlying THQ's antioxidant effects involves the regulation of the Nrf2/HO-1 signaling pathway. Furthermore, THQ has been found to modulate key components of inflammatory signaling pathways, including toll-like receptors (TLRs), nuclear factor-κB (NF-κB), interleukin 6 (IL-6), IL-1β, and tumor necrosis factor alpha (TNFα), thereby exerting anti-inflammatory effects. This comprehensive review explores the various beneficial effects of THQ and its NFs on neurological disorders and provides insights into the underlying mechanisms involved.
Collapse
Affiliation(s)
- Maryam Saadat
- Department of Anatomical Sciences, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Narjes Dahmardeh
- Department of Anatomical Sciences, Faculty of Medicine, Zabol University of Medical Sciences, Zabol, Iran
| | - Fatemeh Sheikhbahaei
- Department of Anatomy, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran.
| | - Tahmineh Mokhtari
- Hubei Key Laboratory of Embryonic Stem Cell Research, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
- Department of Histology and Embryology, Faculty of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| |
Collapse
|
5
|
Burrows K, Figueroa-Hall LK, Stewart JL, Alarbi AM, Kuplicki R, Hannafon BN, Tan C, Risbrough VB, McKinney BA, Ramesh R, Victor TA, Aupperle R, Savitz J, Teague TK, Khalsa SS, Paulus MP. Exploring the role of neuronal-enriched extracellular vesicle miR-93 and interoception in major depressive disorder. Transl Psychiatry 2024; 14:199. [PMID: 38678012 PMCID: PMC11055873 DOI: 10.1038/s41398-024-02907-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 04/02/2024] [Accepted: 04/10/2024] [Indexed: 04/29/2024] Open
Abstract
Major depressive disorder (MDD) is associated with interoceptive processing dysfunctions, but the molecular mechanisms underlying this dysfunction are poorly understood. This study combined brain neuronal-enriched extracellular vesicle (NEEV) technology and serum markers of inflammation and metabolism with Functional Magnetic Resonance Imaging (fMRI) to identify the contribution of gene regulatory pathways, in particular micro-RNA (miR) 93, to interoceptive dysfunction in MDD. Individuals with MDD (n = 41) and healthy comparisons (HC; n = 35) provided blood samples and completed an interoceptive attention task during fMRI. EVs were separated from plasma using a precipitation method. NEEVs were enriched by magnetic streptavidin bead immunocapture utilizing a neural adhesion marker (L1CAM/CD171) biotinylated antibody. The origin of NEEVs was validated with two other neuronal markers - neuronal cell adhesion molecule (NCAM) and ATPase Na+/K+ transporting subunit alpha 3 (ATP1A3). NEEV specificities were confirmed by flow cytometry, western blot, particle size analyzer, and transmission electron microscopy. NEEV small RNAs were purified and sequenced. Results showed that: (1) MDD exhibited lower NEEV miR-93 expression than HC; (2) within MDD but not HC, those individuals with the lowest NEEV miR-93 expression had the highest serum concentrations of interleukin (IL)-1 receptor antagonist, IL-6, tumor necrosis factor, and leptin; and (3) within HC but not MDD, those participants with the highest miR-93 expression showed the strongest bilateral dorsal mid-insula activation during interoceptive versus exteroceptive attention. Since miR-93 is regulated by stress and affects epigenetic modulation by chromatin re-organization, these results suggest that healthy individuals but not MDD participants show an adaptive epigenetic regulation of insular function during interoceptive processing. Future investigations will need to delineate how specific internal and external environmental conditions contribute to miR-93 expression in MDD and what molecular mechanisms alter brain responsivity to body-relevant signals.
Collapse
Affiliation(s)
| | - Leandra K Figueroa-Hall
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health and Natural Sciences, University of Tulsa, Tulsa, OK, USA
| | - Jennifer L Stewart
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health and Natural Sciences, University of Tulsa, Tulsa, OK, USA
| | - Ahlam M Alarbi
- Departments of Surgery and Psychiatry, School of Community Medicine, The University of Oklahoma, Tulsa, OK, USA
| | | | - Bethany N Hannafon
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Chibing Tan
- Departments of Surgery and Psychiatry, School of Community Medicine, The University of Oklahoma, Tulsa, OK, USA
| | - Victoria B Risbrough
- Center of Excellence for Stress and Mental Health, La Jolla, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Brett A McKinney
- Department of Mathematics and Computer Science, University of Tulsa, Tulsa, OK, USA
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Robin Aupperle
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health and Natural Sciences, University of Tulsa, Tulsa, OK, USA
| | - Jonathan Savitz
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health and Natural Sciences, University of Tulsa, Tulsa, OK, USA
| | - T Kent Teague
- Departments of Surgery and Psychiatry, School of Community Medicine, The University of Oklahoma, Tulsa, OK, USA
- Department of Biochemistry and Microbiology, The Oklahoma State University Center for Health Sciences, Tulsa, OK, USA
- Department of Pharmaceutical Sciences, The University of Oklahoma College of Pharmacy, Oklahoma City, OK, USA
| | - Sahib S Khalsa
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health and Natural Sciences, University of Tulsa, Tulsa, OK, USA
| | - Martin P Paulus
- Laureate Institute for Brain Research, Tulsa, OK, USA
- Oxley College of Health and Natural Sciences, University of Tulsa, Tulsa, OK, USA
| |
Collapse
|
6
|
Duan M, Xu Y, Li Y, Feng H, Chen Y. Targeting brain-peripheral immune responses for secondary brain injury after ischemic and hemorrhagic stroke. J Neuroinflammation 2024; 21:102. [PMID: 38637850 PMCID: PMC11025216 DOI: 10.1186/s12974-024-03101-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: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024] Open
Abstract
The notion that the central nervous system is an immunologically immune-exempt organ has changed over the past two decades, with increasing evidence of strong links and interactions between the central nervous system and the peripheral immune system, both in the healthy state and after ischemic and hemorrhagic stroke. Although primary injury after stroke is certainly important, the limited therapeutic efficacy, poor neurological prognosis and high mortality have led researchers to realize that secondary injury and damage may also play important roles in influencing long-term neurological prognosis and mortality and that the neuroinflammatory process in secondary injury is one of the most important influences on disease progression. Here, we summarize the interactions of the central nervous system with the peripheral immune system after ischemic and hemorrhagic stroke, in particular, how the central nervous system activates and recruits peripheral immune components, and we review recent advances in corresponding therapeutic approaches and clinical studies, emphasizing the importance of the role of the peripheral immune system in ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Mingxu Duan
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ya Xu
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yuanshu Li
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Yujie Chen
- Department of Neurosurgery, State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
7
|
Zhang SS, Zhang JW, Zhang KX, Cui WQ, Zhi HW, Li HT, Wu HY, Wang YH. Hsa-miR-877-5p Expression in Acute Ischemic Stroke Based on Bioinformatics Analysis and Clinical Validation. Mol Neurobiol 2024; 61:1990-2005. [PMID: 37837492 DOI: 10.1007/s12035-023-03675-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/24/2023] [Indexed: 10/16/2023]
Abstract
Inflammation and immunity play important roles in the pathogenesis of ischemic stroke. This study aimed to explore key regulatory genes in acute ischemic stroke (AIS) and their underlying mechanisms to provide new research targets for the diagnosis and treatment of ischemic stroke. We searched for differentially expressed mRNAs and miRNAs in patients with AIS and healthy populations in GEO databases, constructed a miRNA-mRNA network, and screened key miRNAs using least absolute shrinkage and selection operator regression and the support vector machine-recursive feature elimination model. Correlations between key miRNAs and infiltrating immune cells and inflammatory factors were analyzed using CIBERSORT and immunoassays and verified using clinical experiments. Bioinformatics analysis identified hsa-miR-877-5p as a key regulatory miRNA in AIS that can modulate immune and inflammatory responses. In clinical studies, it was verified by quantitative PCR analysis that the expression of hsa-miR-877-5p in the blood of AIS patients was higher than that of the healthy group. Then, enzyme-linked immunosorbent assay revealed that the expression of IL-23 and TNF-α related to inflammation in AIS patients was higher than that of the healthy. Quantitative PCR further found that the relative mRNA expression of IL-23, CXCR3, and TNF-α in AIS group was higher than that of the healthy group. This study may provide a basis for a more comprehensive understanding of the potential mechanism of the occurrence and development of AIS, and hsa-miR-877-5p and its downstream effectors IL-23, CXCR3, and TNF-α may be potential intervention targets in AIS.
Collapse
Affiliation(s)
- Si-Shuo Zhang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369 in Lixia District, Jinan, China
| | - Ji-Wei Zhang
- School of Acupuncture-Moxibustion and Tuina, Shandong University of Traditional Chinese Medicine, University Road NO.4655 in Changqing District, Jinan, China
| | - Kai-Xin Zhang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, University Road NO.4655 in Changqing District, Jinan, China
| | - Wen-Qiang Cui
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369 in Lixia District, Jinan, China
| | - Hong-Wei Zhi
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369 in Lixia District, Jinan, China
| | - Hai-Tao Li
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369 in Lixia District, Jinan, China
| | - Hong-Yun Wu
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369 in Lixia District, Jinan, China
| | - Ya-Han Wang
- Department of Neurology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road No.16369 in Lixia District, Jinan, China.
| |
Collapse
|
8
|
He C, Li Z, Yang M, Yu W, Luo R, Zhou J, He J, Chen Q, Song Z, Cheng S. Non-Coding RNA in Microglia Activation and Neuroinflammation in Alzheimer's Disease. J Inflamm Res 2023; 16:4165-4211. [PMID: 37753266 PMCID: PMC10519213 DOI: 10.2147/jir.s422114] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by complex pathophysiological features. Amyloid plaques resulting from extracellular amyloid deposition and neurofibrillary tangles formed by intracellular hyperphosphorylated tau accumulation serve as primary neuropathological criteria for AD diagnosis. The activation of microglia has been closely associated with these pathological manifestations. Non-coding RNA (ncRNA), a versatile molecule involved in various cellular functions such as genetic information storage and transport, as well as catalysis of biochemical reactions, plays a crucial role in microglial activation. This review aims to investigate the regulatory role of ncRNAs in protein expression by directly targeting genes, proteins, and interactions. Furthermore, it explores the ability of ncRNAs to modulate inflammatory pathways, influence the expression of inflammatory factors, and regulate microglia activation, all of which contribute to neuroinflammation and AD. However, there are still significant controversies surrounding microglial activation and polarization. The categorization into M1 and M2 phenotypes may oversimplify the intricate and multifaceted regulatory processes in microglial response to neuroinflammation. Limited research has been conducted on the role of ncRNAs in regulating microglial activation and inducing distinct polarization states in the context of neuroinflammation. Moreover, the regulatory mechanisms through which ncRNAs govern microglial function continue to be refined. The current understanding of ncRNA regulatory pathways involved in microglial activation remains incomplete and may be influenced by spatial, temporal, and tissue-specific factors. Therefore, further in-depth investigations are warranted. In conclusion, there are ongoing debates and uncertainties regarding the activation and polarization of microglial cells, particularly concerning the categorization into M1 and M2 phenotypes. The study of ncRNA regulation in microglial activation and polarization, as well as its mechanisms, is still in its early stages and requires further investigation. However, this review offers new insights and opportunities for therapeutic approaches in AD. The development of ncRNA-based drugs may hold promise as a new direction in AD treatment.
Collapse
Affiliation(s)
- Chunxiang He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Ze Li
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Miao Yang
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Wenjing Yu
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Rongsiqing Luo
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Jinyong Zhou
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Jiawei He
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Qi Chen
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Zhenyan Song
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Shaowu Cheng
- School of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
9
|
Burrows K, Figueroa-Hall L, Stewart J, Alarbi A, Kuplicki R, Hannafon B, Tan C, Risbrough V, McKinney B, Ramesh R, Victor T, Aupperle R, Savitz J, Teague K, Khalsa S, Paulus M. Exploring the role of neuronal-enriched extracellular vesicle miR-93 and interoception in major depressive disorder. RESEARCH SQUARE 2023:rs.3.rs-2813878. [PMID: 37398092 PMCID: PMC10312986 DOI: 10.21203/rs.3.rs-2813878/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Major depressive disorder (MDD) is associated with interoceptive processing dysfunctions, but the molecular mechanisms underlying this dysfunction are poorly understood. This study combined brain Neuronal-Enriched Extracellular Vesicle (NEEV) technology and serum markers of inflammation and metabolism with Functional Magnetic Resonance Imaging (fMRI) to identify the contribution of gene regulatory pathways, in particular micro-RNA (miR) 93, to interoceptive dysfunction in MDD. Individuals with MDD (n = 44) and healthy comparisons (HC; n = 35) provided blood samples and completed an interoceptive attention task during fMRI. EVs were separated from plasma using a precipitation method. NEEVs were enriched by magnetic streptavidin bead immunocapture utilizing a neural adhesion marker (CD171) biotinylated antibody. NEEV specificities were confirmed by ow cytometry, western blot, particle size analyzer, and transmission electron microscopy. NEEV small RNAs were purified and sequenced. Results showed that: (1) MDD exhibited lower NEEV miR-93 expression than HC; (2) within MDD but not HC, those individuals with the lowest NEEV miR-93 expression had the highest serum concentrations of interleukin (IL)-1 receptor antagonist, IL-6, tumor necrosis factor, and leptin; and (3) within HC but not MDD, those participants with the highest miR-93 expression showed the strongest bilateral dorsal mid-insula activation. Since miR-93 is regulated by stress and affects epigenetic modulation by chromatin reorganization, these results suggest that healthy individuals but not MDD participants show an adaptive epigenetic regulation of insular function during interoceptive processing. Future investigations will need to delineate how specific internal and external environmental conditions contribute to miR-93 expression in MDD and what molecular mechanisms alter brain responsivity to body-relevant signals.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Kent Teague
- University of Oklahoma School of Community Medicine
| | | | | |
Collapse
|
10
|
Zhang J, Ma H, Yang G, Ke J, Sun W, Yang L, Kuang S, Li H, Yuan W. Differentially expressed miRNA profiles of serum-derived exosomes in patients with sudden sensorineural hearing loss. Front Neurol 2023; 14:1177988. [PMID: 37332997 PMCID: PMC10273844 DOI: 10.3389/fneur.2023.1177988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/10/2023] [Indexed: 06/20/2023] Open
Abstract
Objectives This study aimed to compare the expressed microRNA (miRNA) profiles of serum-derived exosomes of patients with sudden sensorineural hearing loss (SSNHL) and normal hearing controls to identify exosomal miRNAs that may be associated with SSNHL or serve as biomarkers for SSNHL. Methods Peripheral venous blood of patients with SSNHL and healthy controls was collected to isolate exosomes. Nanoparticle tracking analysis, transmission electron microscopy, and Western blotting were used to identify the isolated exosomes, after which total RNA was extracted and used for miRNA transcriptome sequencing. Differentially expressed miRNAs (DE-miRNAs) were identified based on the thresholds of P < 0.05 and |log2fold change| > 1 and subjected to functional analyses. Finally, four exosomal DE-miRNAs, including PC-5p-38556_39, PC-5p-29163_54, PC-5p-31742_49, and hsa-miR-93-3p_R+1, were chosen for validation using quantitative real-time polymerase chain reaction (RT-qPCR). Results Exosomes were isolated from serum and identified based on particle size, morphological examination, and expression of exosome-marker proteins. A total of 18 exosomal DE-miRNAs, including three upregulated and 15 downregulated miRNAs, were found in SSNHL cases. Gene ontology (GO) functional annotation analysis revealed that target genes in the top 20 terms were mainly related to "protein binding," "metal ion binding," "ATP binding," and "intracellular signal transduction." Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis revealed that these target genes were functionally enriched in the "Ras," "Hippo," "cGMP-PKG," and "AMPK signaling pathways." The expression levels of PC-5p-38556_39 and PC-5p-29163_54 were significantly downregulated and that of miR-93-3p_R+1 was highly upregulated in SSNHL. Consequently, the consistency rate between sequencing and RT-qPCR was 75% and sequencing results were highly reliable. Conclusion This study identified 18 exosomal DE-miRNAs, including PC-5p-38556_39, PC-5p-29163_54, and miR-93-3p, which may be closely related to SSNHL pathogenesis or serve as biomarkers for SSNHL.
Collapse
Affiliation(s)
- Juhong Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
- School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Haizhu Ma
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
| | - Guijun Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
| | - Jing Ke
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
- School of Basic Medicine, Chongqing Medical University, Chongqing, China
| | - Wenfang Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
| | - Li Yang
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
| | - Shaojing Kuang
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
| | - Hai Li
- Department of Otorhinolaryngology Head and Neck Surgery, Xuanhan County People's Hospital, Dazhou, Sichuan, China
| | - Wei Yuan
- Department of Otorhinolaryngology Head and Neck Surgery, Chongqing General Hospital, Chongqing, China
- School of Basic Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Qi D, Wang W, Zhang Y, Zhang T. MiR-99b regulates cerebral ischemia neuronal injury through targeting IGF1R. Panminerva Med 2023; 65:30-36. [PMID: 32343508 DOI: 10.23736/s0031-0808.20.03920-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND Recently, microRNA-99b (miR-99b) shows diverse functions in different human disease. However, further studies about the potential effect of miR-99b in cerebral ischemia injury still need to be done. METHODS The expressions of miR-99b and IGF1R were detected via RT-qPCR assay. Western blot assay was applied to measure the protein expression of Caspase-3, Bax and Bcl-2. MTT assay was used to observe cell viability of SH-SY5Y cells. The association of miR-99b and IGF1R was testified by dual luciferase assay. And human SH-SY5Y cells were treated with the oxygen-glucose deprivation/reperfusion (OGD/R) to mimic CIR injury. RESULTS The expression of miR-99b was increased in the OGD/R model. And upregulation of miR-99b promoted cell viability and inhibited apoptosis induced by OGD/R. Moreover, IGF1R was confirmed as a direct target gene of miR-99b. The expression of IGF1R was obviously decreased under OGD/R conditions. CONCLUSIONS MiR-99b promoted the viability and suppressed apoptosis of SH-SY5Y cells under OGD/R conditions through targeting IGF1R.
Collapse
Affiliation(s)
- Dengbin Qi
- Department of Neurology, Affiliated Hospital of Jining Medical University, YanZhou Branch, Jining, China
| | - Wei Wang
- Disinfection Supply Center, Qingdao Municipal Hospital, Qingdao, China
| | - Ying Zhang
- Department of Internal Medicine, Binzhou People's Hospital, Binzhou, China
| | - Tao Zhang
- Department of Cardiovascular Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China -
| |
Collapse
|
12
|
Yang K, Zeng L, Ge A, Wang S, Zeng J, Yuan X, Mei Z, Wang G, Ge J. A systematic review of the research progress of non-coding RNA in neuroinflammation and immune regulation in cerebral infarction/ischemia-reperfusion injury. Front Immunol 2022; 13:930171. [PMID: 36275741 PMCID: PMC9585453 DOI: 10.3389/fimmu.2022.930171] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/08/2022] [Indexed: 11/15/2022] Open
Abstract
Cerebral infarction/ischemia-reperfusion injury is currently the disease with the highest mortality and disability rate of cardiovascular disease. Current studies have shown that nerve cells die of ischemia several hours after ischemic stroke, which activates the innate immune response in the brain, promotes the production of neurotoxic substances such as inflammatory cytokines, chemokines, reactive oxygen species and − nitrogen oxide, and mediates the destruction of blood-brain barrier and the occurrence of a series of inflammatory cascade reactions. Meanwhile, the expression of adhesion molecules in cerebral vascular endothelial cells increased, and immune inflammatory cells such as polymorphonuclear neutrophils, lymphocytes and mononuclear macrophages passed through vascular endothelial cells and entered the brain tissue. These cells recognize antigens exposed by the central nervous system in the brain, activate adaptive immune responses, and further mediate secondary neuronal damage, aggravating neurological deficits. In order to reduce the above-mentioned damage, the body induces peripheral immunosuppressive responses through negative feedback, which increases the incidence of post-stroke infection. This process is accompanied by changes in the immune status of the ischemic brain tissue in local and systemic systems. A growing number of studies implicate noncoding RNAs (ncRNAs) as novel epigenetic regulatory elements in the dysfunction of various cell subsets in the neurovascular unit after cerebral infarction/ischemia-reperfusion injury. In particular, recent studies have revealed advances in ncRNA biology that greatly expand the understanding of epigenetic regulation of immune responses and inflammation after cerebral infarction/ischemia-reperfusion injury. Identification of aberrant expression patterns and associated biological effects of ncRNAs in patients revealed their potential as novel biomarkers and therapeutic targets for cerebral infarction/ischemia-reperfusion injury. Therefore, this review systematically presents recent studies on the involvement of ncRNAs in cerebral infarction/ischemia-reperfusion injury and neuroimmune inflammatory cascades, and elucidates the functions and mechanisms of cerebral infarction/ischemia-reperfusion-related ncRNAs, providing new opportunities for the discovery of disease biomarkers and targeted therapy. Furthermore, this review introduces clustered regularly interspaced short palindromic repeats (CRISPR)-Display as a possible transformative tool for studying lncRNAs. In the future, ncRNA is expected to be used as a target for diagnosing cerebral infarction/ischemia-reperfusion injury, judging its prognosis and treatment, thereby significantly improving the prognosis of patients.
Collapse
Affiliation(s)
- Kailin Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Liuting Zeng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Anqi Ge
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Shanshan Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinsong Zeng
- The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Xiao Yuan
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Guozuo Wang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, China
- Hunan Academy of Chinese Medicine, Changsha, China
- *Correspondence: Jinwen Ge,
| |
Collapse
|
13
|
Luo H, Huang F, Huang Z, Huang H, Liu C, Feng Y, Qi Z. microRNA-93 packaged in extracellular vesicles from mesenchymal stem cells reduce neonatal hypoxic-ischemic brain injury. Brain Res 2022; 1794:148042. [PMID: 35952773 DOI: 10.1016/j.brainres.2022.148042] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/31/2022] [Accepted: 08/05/2022] [Indexed: 01/08/2023]
Abstract
OBJECTIVE Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) have been proposed as a promising strategy for treating ischemia-related diseases. Herein, we probed into the role of miR-93 delivered by BMSC-EVs in hypoxic-ischemic brain injury (HIBD). METHODS Neonatal HIBD mouse models and hippocampal neuron models of oxygen glucose deprivation (OGD) were constructed. EVs were isolated from the culture medium of bone marrow MSCs (BMSCs). After co-culture of BMSC-EVs with OGD-exposed hippocampal neurons, the effect of microRNA-93 (miR-93) delivered by BMSC-EVs on OGD-induced hippocampal neurons as well as on HIBD in vivo under transfection of miR-93 mimic or inhibitor was explored. The interaction among miR-93, JMJD3, and p53/KLF2 axis was assessed. RESULTS BMSC-EVs prevented OGD-induced hippocampal neuron apoptosis and inflammation, which was associated with their transfer of miR-93 into the hippocampal neurons. miR-93 targeted JMJD3 and downregulated its expression, thus inhibiting the OGD-induced hippocampal neuron apoptosis. By regulating the JMJD3/p53/KLF2 axis, miR-93 in BMSC-EVs reduced the OGD-induced hippocampal neuron apoptosis in vitro as well as alleviating HIBD in vivo. CONCLUSIONS The current study highlighted that miR-93 delivered by BMSC-EVs alleviated HIBD in neonatal mice through the JMJD3-dependent p53/KLF2 axis.
Collapse
Affiliation(s)
- Hongcheng Luo
- Department of Medical Laboratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China; Medical College of Guangxi University, Nanning 530004, Guangxi, China
| | - Fugao Huang
- Department of Ultrasound, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Zhijing Huang
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China
| | - Huatuo Huang
- Department of Medical Laboratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Chunhong Liu
- Department of Medical Laboratory, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Yanni Feng
- Department of Pediatrics, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, China.
| | - Zhongquan Qi
- Medical College of Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
14
|
Shi X, Zhong X, Deng L, Wu X, Zhang P, Zhang X, Wang G. Mesenchymal stem cell-derived extracellular vesicle-enclosed miR-93 prevents hypoxic-ischemic brain damage in rats. Neuroscience 2022; 500:12-25. [PMID: 35803492 DOI: 10.1016/j.neuroscience.2022.06.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 06/24/2022] [Accepted: 06/27/2022] [Indexed: 02/06/2023]
Abstract
Hypoxic-ischemic brain damage (HIBD) usually induces chronic neurological disorder and even acute death, but effective neuroprotective strategy is still limited. Herein, we performed this study to clarify the mechanism of mesenchymal stem cell (MSC)-derived extracellular vesicles (EVs) containing microRNA-93 (miR-93) in influencing this damage via regulation of the histone deacetylase 4 (HDAC4)/B-cell lymphoma-2 (Bcl-2) axis. Initially, differentially expressed Bcl-2 was identified in middle cerebral artery occlusion (MCAO), and the upstream regulatory miR-93 and its potential target HDAC4 were also predicted through bioinformatics analysis. HIBD was modeled in vitro by exposing hippocampal neurons to oxygen-glucose deprivation (OGD) and in vivo by MCAO in rats. EVs were isolated from the bone marrow MSCs of well-grown rats. Our experimental data validated that HDAC4 was highly expressed while miR-93 and Bcl-2 were poorly expressed in MCAO rats. Furthermore, HDAC4 overexpression, through inhibiting Bcl-2 via deacetylation, promoted the infarct volume and pathological changes in hippocampal tissues and neuron apoptosis, and impaired neurobehavioral ability of MCAO rats. Of note, miR-93 was found to target HDAC4. Importantly, MSC-derived EVs overexpressing miR-93 suppressed HDAC4 expression and subsequently impeded the apoptosis of OGD-exposed hippocampal neurons in vitro, and also ameliorated HIBD in vivo. Taken together, miR-93 delivered by MSC-derived EVs can ameliorate HIBD by suppressing hippocampal neuron apoptosis through targeting the HDAC4/Bcl-2 axis, a finding which may be of great significance in the treatment of HIBD.
Collapse
Affiliation(s)
- Xiaoding Shi
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150081, P. R. China
| | - Xuelai Zhong
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150081, P. R. China
| | - Lin Deng
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150081, P. R. China
| | - Xiaohong Wu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150081, P. R. China
| | - Pinyi Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150081, P. R. China
| | - Xin Zhang
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Harbin 150081, P. R. China
| | - Guonian Wang
- Department of Anesthesiology, The Fourth Hospital of Harbin Medical University, Harbin 150001, P. R. China.
| |
Collapse
|
15
|
Xu X, Zhi T, Hua L, Jiang K, Chen C. IRAK4 exacerbates traumatic brain injury via activation of TAK1 signaling pathway. Exp Neurol 2022; 351:114007. [PMID: 35149117 DOI: 10.1016/j.expneurol.2022.114007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 01/29/2022] [Accepted: 02/04/2022] [Indexed: 11/04/2022]
Abstract
Although multiple signaling pathways contributing to the pathophysiological process have been investigated, treatments for traumatic brain injury (TBI) against present targets have not acquired significant clinical progress. Interleukin-1 receptor-associated kinase 4 (IRAK4) is an important factor involved in regulating immunity and inflammation. However, the role of IRAK4 in TBI still remains largely unknown. Therefore, using a controlled cortical impact model (CCI), we investigated the function and molecular mechanism of IRAK4 in the context of TBI. IRAK4 was found to be activated in a time-dependent manner after TBI and mainly expressed in neurons. Inhibition of IRAK4 by siRNAs could significantly alleviates neuroinflammation, neuron apoptosis, brain edema, brain-blood barrier (BBB) dysfunction and improves neurological deficit in the context of CCI. Mechanistically, IRAK4 exacerbates CCI via activation of TAK1 signaling pathway. Interestingly, PF-0665083, an IRAK4 inhibitor, inhibits phosphorylation of IRAK4 and attenuates CCI-induced secondary injury. It could be conclude that IRAK4 plays a critical role in TBI-induced secondary injury and the underlining mechanism may be related to activation of TAK1 signaling pathway. PF-0665083 may serve as a potential treatment strategy to relieve TBI.
Collapse
Affiliation(s)
- Xiupeng Xu
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu Province, China
| | - Tongle Zhi
- Department of Neurosurgery, The First People's Hospital of Yancheng, the Fourth Affiliated Hospital of Nantong University, Yancheng 224006, Jiangsu Province, China
| | - Lingyang Hua
- Department of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai 200000, China
| | - Kuan Jiang
- Department of Neurosurgery, Yixing People's Hospital, Yixing 214200, Jiangsu Province, China
| | - Chen Chen
- Department of Cardiology, Nanjing Brain Hospital Affiliated to Nanjing Medical University, Nanjing 210029, Jiangsu Province, China.
| |
Collapse
|
16
|
Penning A, Tosoni G, Abiega O, Bielefeld P, Gasperini C, De Pietri Tonelli D, Fitzsimons CP, Salta E. Adult Neural Stem Cell Regulation by Small Non-coding RNAs: Physiological Significance and Pathological Implications. Front Cell Neurosci 2022; 15:781434. [PMID: 35058752 PMCID: PMC8764185 DOI: 10.3389/fncel.2021.781434] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/09/2021] [Indexed: 01/11/2023] Open
Abstract
The adult neurogenic niches are complex multicellular systems, receiving regulatory input from a multitude of intracellular, juxtacrine, and paracrine signals and biological pathways. Within the niches, adult neural stem cells (aNSCs) generate astrocytic and neuronal progeny, with the latter predominating in physiological conditions. The new neurons generated from this neurogenic process are functionally linked to memory, cognition, and mood regulation, while much less is known about the functional contribution of aNSC-derived newborn astrocytes and adult-born oligodendrocytes. Accumulating evidence suggests that the deregulation of aNSCs and their progeny can impact, or can be impacted by, aging and several brain pathologies, including neurodevelopmental and mood disorders, neurodegenerative diseases, and also by insults, such as epileptic seizures, stroke, or traumatic brain injury. Hence, understanding the regulatory underpinnings of aNSC activation, differentiation, and fate commitment could help identify novel therapeutic avenues for a series of pathological conditions. Over the last two decades, small non-coding RNAs (sncRNAs) have emerged as key regulators of NSC fate determination in the adult neurogenic niches. In this review, we synthesize prior knowledge on how sncRNAs, such as microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs), may impact NSC fate determination in the adult brain and we critically assess the functional significance of these events. We discuss the concepts that emerge from these examples and how they could be used to provide a framework for considering aNSC (de)regulation in the pathogenesis and treatment of neurological diseases.
Collapse
Affiliation(s)
- Amber Penning
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Giorgia Tosoni
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Caterina Gasperini
- Neurobiology of miRNAs Lab, Istituto Italiano di Tecnologia, Genova, Italy
| | | | - Carlos P. Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, Netherlands
| | - Evgenia Salta
- Laboratory of Neurogenesis and Neurodegeneration, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
17
|
Liu J, Ai P, Sun Y, Yang X, Li C, Liu Y, Xia X, Zheng JC. Propofol Inhibits Microglial Activation via miR-106b/Pi3k/Akt Axis. Front Cell Neurosci 2021; 15:768364. [PMID: 34776870 PMCID: PMC8581742 DOI: 10.3389/fncel.2021.768364] [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: 08/31/2021] [Accepted: 10/06/2021] [Indexed: 12/05/2022] Open
Abstract
Propofol is an established intravenous anesthetic agent with potential neuroprotective effects. In this study, we investigated the roles and the underlying mechanisms of propofol in inhibiting the pro-inflammatory responses of microglia. Propofol significantly reduced the messenger RNA (mRNA) levels of Tnf, Nos2, and NF-κB pathway related genes Ticam1, Myd88, Irf3, and Nfkb1 in lipopolysaccharide (LPS)-treated primary microglia. After screening the miRNA profiles in microglia under LPS and propofol treatment conditions, we found propofol abrogated the LPS-induced misexpression of miRNAs including miR-106b, miR-124, miR-185, and miR-9. Perturbation of function approaches suggested miR-106b as the core miRNA that mediated the anti-inflammatory effects of propofol on microglial activation. RNA sequencing (RNA-seq) analysis further identified Pi3k/Akt signaling as one of the most affected pathways after miR-106b perturbation of function. The treatment of Pi3k/Akt signaling agonist 740Y-P elevated miR-106b-reduced Akt phosphorylation and abolished the inhibitory effects of miR-106b on the pro-inflammatory responses of microglia. Our results suggest propofol inhibits microglial activation via miR-106b/Pi3k/Akt axis, shedding light on a novel molecular mechanism of propofol-mediated immunomodulatory effects and implying propofol as potential therapeutics for treating neuroinflammation-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Pu Ai
- Wuxi Clinical College of Anhui Medical University, Hefei, China
| | - Yiyan Sun
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaoyu Yang
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Chunhong Li
- Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yihan Liu
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaohuan Xia
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jialin C Zheng
- Department of Anesthesiology, Tongji Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital Affiliated to Tongji University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| |
Collapse
|
18
|
Moradi Z, Rabiei Z, Anjomshoa M, Amini-Farsani Z, Massahzadeh V, Asgharzade S. Neuroprotective effect of wild lowbush blueberry (Vaccinium angustifolium) on global cerebral ischemia/reperfusion injury in rats: Downregulation of iNOS/TNF-α and upregulation of miR-146a/miR-21 expression. Phytother Res 2021; 35:6428-6440. [PMID: 34580912 DOI: 10.1002/ptr.7296] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 07/23/2021] [Accepted: 09/08/2021] [Indexed: 11/08/2022]
Abstract
This study aims to investigate the neuroprotective effect of wild lowbush blueberry on CIRI in rats. Accordingly, CIRI and reperfusion were induced in rats for 60 min and 24 h, respectively. Then, the mechanisms of the neuroprotective effects of BBE were investigated in the injury through evaluating miR-146a, miR-21, and their targets in a CIRI rat model. After that, the BBE (30, 60, and 120 mg/kg b.wt) was intraperitoneally injected for 14 days, then CIRI was induced by BCCAO for 60 min for ischemic stroke and reperfusion for 24 h. Several parameters including the oxidative stress levels in the hippocampus and serum were measured 24 h after the CIRI. The findings showed that the BBE significantly decreased the levels of malondialdehyde (MDA) and nitric oxide (NO) and increased ferric ion reducing antioxidant power (FRAP) levels in the hippocampus and serum following the stroke. The BBE also maximized the miR-146a and miR-21 expressions and moderated iNOS and TNF-α expressions in the hippocampus. Likewise, the BBE enlarged the CA1 and CA3 domains of the post-stroke pyramidal cell layers of the hippocampus. Overall, the results revealed that BBE had potent neuroprotective efficacy against CIRI via the effective modulation of neuroinflammatory cascades and protected neurons against ischemic death.
Collapse
Affiliation(s)
- Zahra Moradi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zahra Rabiei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Maryam Anjomshoa
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Zeinab Amini-Farsani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Vahid Massahzadeh
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
19
|
Muthusami S, Ramachandran I, Krishnamoorthy S, Sambandam Y, Ramalingam S, Queimado L, Chaudhuri G, Ramachandran IK. Regulation of MicroRNAs in Inflammation-Associated Colorectal Cancer: A Mechanistic Approach. Endocr Metab Immune Disord Drug Targets 2021; 21:67-76. [PMID: 32940190 DOI: 10.2174/1871530320666200917112802] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/29/2020] [Accepted: 04/29/2020] [Indexed: 11/22/2022]
Abstract
The development of colorectal cancer (CRC) is a multistage process. The inflammation of
the colon as in inflammatory bowel disease (IBD) such as ulcerative colitis (UC) or Crohn’s disease
(CD) is often regarded as the initial trigger for the development of inflammation-associated CRC.
Many cytokines such as tumor necrosis factor alpha (TNF-α) and interleukins (ILs) are known to exert
proinflammatory actions, and inflammation initiates or promotes tumorigenesis of various cancers,
including CRC, through differential regulation of microRNAs (miRNAs/miRs). miRNAs can be
oncogenic miRNAs (oncomiRs) or anti-oncomiRs/tumor suppressor miRNAs, and they play key roles
during colorectal carcinogenesis. However, the functions and molecular mechanisms of regulation of
miRNAs involved in inflammation-associated CRC are still anecdotal and largely unknown.
Consolidating the published results and offering perspective solutions to circumvent CRC, the current
review is focused on the role of miRNAs and their regulation in the development of CRC. We have
also discussed the model systems adapted by researchers to delineate the role of miRNAs in
inflammation-associated CRC.
Collapse
Affiliation(s)
- Sridhar Muthusami
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Ilangovan Ramachandran
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai 600 113, Tamil Nadu, India
| | - Sneha Krishnamoorthy
- Department of Biochemistry, Karpagam Academy of Higher Education, Coimbatore 641 021, Tamil Nadu, India
| | - Yuvaraj Sambandam
- Department of Surgery, Comprehensive Transplant Center, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, United States
| | - Satish Ramalingam
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Kanchipuram 603 203, Tamil Nadu, India
| | - Lurdes Queimado
- Departments of Otorhinolaryngology - Head and Neck Surgery, Cell Biology, Pediatrics, The University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, United States
| | - Gautam Chaudhuri
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, United States
| | | |
Collapse
|
20
|
MicroRNA-93/STAT3 signalling pathway mediates retinal microglial activation and protects retinal ganglion cells in an acute ocular hypertension model. Cell Death Dis 2021; 12:41. [PMID: 33414426 PMCID: PMC7791106 DOI: 10.1038/s41419-020-03337-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 11/28/2020] [Accepted: 12/09/2020] [Indexed: 01/29/2023]
Abstract
Glaucoma is a common neurodegenerative disease and a leading cause of irreversible blindness worldwide. Retinal microglia-mediated neuroinflammation is involved in the process of optic nerve damage, but the mechanisms driving this microglial activation remain mostly elusive. Previous investigations reported that microRNAs are associated with the retinal microglial reaction and neural apoptosis. In the present study, we found that microRNA-93-5p (miR-93) played a key role in the reaction of retinal microglial cells in vivo and in vitro. The miR-93 level was significantly reduced in the retinae of rat acute ocular hypertension (AOH) models, which were accompanied by retinal microglial activation, overproduction of inflammatory cytokines, and subsequent retinal ganglion cells (RGCs) death, versus the retinae of controls. The induction of miR-93 overexpression significantly reduced microglial proliferation, migration and cytokine release, inhibited the expression of the target gene signal transducer and activator of transcription 3 (STAT3) and p-STAT3, and was associated with a reduced loss of RGCs. Treatment with a STAT3 inhibitor also decreased retinal microglial activation after AOH injury. Taken together, these results suggest that the miR-93/STAT3 pathway is directly related to the downregulation of retinal microglia-mediated neuro-inflammation and showed a neuroprotective effect. Regulating microglial activation through miR-93 may serve as a target for neuroprotective therapy in pathological ocular hypertension.
Collapse
|
21
|
Xu H, Nie B, Liu L, Zhang C, Zhang Z, Xu M, Mei Y. Curcumin Prevents Brain Damage and Cognitive Dysfunction During Ischemic-reperfusion Through the Regulation of miR-7-5p. Curr Neurovasc Res 2020; 16:441-454. [PMID: 31660818 DOI: 10.2174/1567202616666191029113633] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/23/2019] [Accepted: 09/28/2019] [Indexed: 12/13/2022]
Abstract
OBJECTIVE This study was to investigate the potential protective effects of curcumin in cerebral ischemia-reperfusion (CIR) and its regulation of miR-7. METHODS Rats were occluded by middle cerebral artery occlusion (MCAO) for 1.5 h and reperfused for 2 h to establish a local CIR model. After 24 hours of model establishment, MCAO rats were given curcumin for 3 days by intragastric administration. PC12 cells were cultured for 6 h in oxygen-glucose deprivation medium and then reoxygenated for 24 h to establish an oxygenglucose deprivation/reoxygenation (OGD/R) model. The OGD/R model cells were treated with curcumin for 48 h. RESULTS Curcumin inhibited the decrease of miR-7-5p expression and an increase of RelA p65 expression induced by CIR and ODG/R. RelA p65 was a target of miR-7-5p. MiR-7-5p antagonists were able to counteract the effect of curcumin on the expression of RelA p65 in ischemic brain tissue of MCAO rats and OGD/R model cells. Curcumin improved OGD/R-induced inhibition of cell activity, necrosis and apoptosis. Curcumin significantly reduced the levels of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1β, reactive oxygen species (ROS) and malondialdehyde (MDA) and increased the activity of superoxide dismutases (SOD) and catalase (CAT) in OGD/R-induced cells. Curcumin may inhibit OGD/R-induced cell damage by regulating miR-7-5p. Curcumin improved cerebral infarction, nerve damage and cognitive dysfunction in rats with CIR, which may be related to the regulation of miR-7-5p/RelA p65 axis. CONCLUSION Curcumin exerts cerebral protection by attenuating cell necrosis and apoptosis, inflammatory response and oxidative stress following CIR, which may be related to its regulation of the miR-7/RELA p65 axis.
Collapse
Affiliation(s)
- Hui Xu
- Department of Fundamental Nursing, The Nursing & Health College of Zhengzhou University, Zhengzhou City, 450001, China
| | - Beibei Nie
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450000, China
| | - Lamei Liu
- Department of Clinical Nursing, The Nursing & Health College of Zhengzhou University, Zhengzhou City, 450001, China
| | - Chunhui Zhang
- Department of Clinical Nursing, The Nursing & Health College of Zhengzhou University, Zhengzhou City, 450001, China
| | - Zhenxiang Zhang
- Department of Fundamental Nursing, The Nursing & Health College of Zhengzhou University, Zhengzhou City, 450001, China
| | - Mengya Xu
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou City, 450000, China
| | - Yongxia Mei
- Department of Public Nursing, The Nursing & Health College of Zhengzhou University, Zhengzhou City, 450001, China
| |
Collapse
|
22
|
Gao H, Xiao D, Gao L, Li X. MicroRNA‑93 contributes to the suppression of lung inflammatory responses in LPS‑induced acute lung injury in mice via the TLR4/MyD88/NF‑κB signaling pathway. Int J Mol Med 2020; 46:561-570. [PMID: 32468034 PMCID: PMC7307825 DOI: 10.3892/ijmm.2020.4610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 03/19/2020] [Indexed: 12/12/2022] Open
Abstract
Acute lung injury (ALI) is a severe inflammatory lung disease with a rapid onset. The anti-inflammatory functions of microRNA-93 (miRNA/miR-93) have been described in various types of tissue injury and disease. However, the biological role of miR-93 and its molecular mechanisms underlying the initiation and progression of ALI have not yet been reported, at least to the best of our knowledge. The present study aimed to investigate the regulatory effects exerted by miR-93 in ALI. Using an in vivo murine model of ALI induced by lipopolysaccharide (LPS), miR-93 expression was found to be downregulated in the lung tissues and bronchoalveolar lavage fluid (BALF) compared with the control group. Following agomiR-93 injection, it was observed that agomiR-93 attenuated lung injury, as evidenced by decreased lung permeability, a reduced lung wet/dry weight ratio and an increased survival rate of the mice. Concomitantly, agomiR-93 significantly reduced LPS-induced the interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α levels in BALF. Of note, Toll-like receptor 4 (TLR4), an upstream regulator of the nuclear factor (NF)-κB signaling pathway, was directly suppressed by miR-93 in RAW 264.7 cells. Importantly, agomiR-93 induced a significant suppression of the TLR4/myeloid differentiation primary response 88 (MyD88)/NF-κB signaling pathway, as demonstrated by the downregulation of MyD88, and the phosphorylation of IκB-α and p65 in the lung tissues of mice with ALI. Taken together, the findings of the present study indicate that miR-93 attenutes LPS-induced lung injury by regulating the TLR4/MyD88/NF-κB signaling pathway, suggesting that miR-93 may prove to be a potential therapeutic target for ALI.
Collapse
Affiliation(s)
- Hu Gao
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| | - Dongqiong Xiao
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| | - Linbo Gao
- Laboratory of Molecular Translational Medicine, Center for Translational Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| | - Xihong Li
- Department of Emergency, West China Second University Hospital, Sichuan University, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
23
|
Hübner M, Moellhoff N, Effinger D, Hinske CL, Hirschberger S, Wu T, Müller MB, Strauß G, Kreth FW, Kreth S. MicroRNA-93 acts as an "anti-inflammatory tumor suppressor" in glioblastoma. Neurooncol Adv 2020; 2:vdaa047. [PMID: 32642700 PMCID: PMC7282490 DOI: 10.1093/noajnl/vdaa047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Background Inflammation is an important driver of malignant glioma disease. Inflammatory mediators are not only produced by immune cells in the tumor microenvironment, but also by glioblastoma (GBM) cells themselves creating a mutually reinforcing loop. We here aimed at identifying an “anti-inflammatory switch” that allows to dampen inflammation in GBM. Methods We used human GBM specimens, primary cultures, and cell lines. The response of GBM cells toward inflammatory stimuli was tested by incubation with supernatant of stimulated human immune cells. Expression levels were measured by whole transcriptome microarrays and qRT-PCR, and protein was quantified by LUMINEX and SDS-PAGE. MicroRNA binding to 3′UTRs was analyzed by luciferase assays. Proliferation rates were determined by flow cytometry, and invasion and angiogenesis were studied using migration and endothelial tube formation assays. Results We demonstrated GBM cells to secrete high amounts of proinflammatory mediators in an inflammatory microenvironment. We found miR-93 as a potential “anti-inflammatory tumor suppressor” dramatically downregulated in GBM. Concordantly, cytokine secretion dropped after miR-93 re-expression. Transfection of miR-93 in GBM cells led to down-regulation of hubs of the inflammatory networks, namely, HIF-1α and MAP3K2 as well as IL-6, G-CSF, IL-8, LIF, IL-1β, COX2, and CXCL5. We showed only COX2 and CXCL5 to be indirectly regulated by miR-93 while all other genes are true targets. Phenotypically, re-expression of miR-93 in GBM cells substantially suppressed proliferation, migration, and angiogenesis. Conclusions Alleviating GBM-derived inflammation by re-expression of miR-93 may be a powerful tool to mitigate these tumors’ aggressiveness and holds promise for new clinical approaches.
Collapse
Affiliation(s)
- Max Hübner
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Nicholas Moellhoff
- Division of Hand, Plastic and Aesthetic Surgery, University Hospital, LMU Munich, Munich, Germany
| | - David Effinger
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Simon Hirschberger
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Tingting Wu
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany
| | - Martin Bernhard Müller
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | - Gabriele Strauß
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| | | | - Simone Kreth
- Walter-Brendel Center of Experimental Medicine, Faculty of Medicine, LMU Munich, Munich, Germany.,Department of Anesthesiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
24
|
Plachel F, Heuberer P, Gehwolf R, Frank J, Tempfer H, Lehner C, Weissenbacher N, Wagner A, Weigl M, Moroder P, Hackl M, Traweger A. MicroRNA Profiling Reveals Distinct Signatures in Degenerative Rotator Cuff Pathologies. J Orthop Res 2020; 38:202-211. [PMID: 31520478 PMCID: PMC6973295 DOI: 10.1002/jor.24473] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 09/03/2019] [Indexed: 02/04/2023]
Abstract
MicroRNAs (miRNAs) have emerged as key regulators orchestrating a wide range of inflammatory and fibrotic diseases. However, the role of miRNAs in degenerative shoulder joint disorders is poorly understood. The aim of this explorative case-control study was to identify pathology-related, circulating miRNAs in patients with chronic rotator cuff tendinopathy and degenerative rotator cuff tears (RCT). In 2017, 15 patients were prospectively enrolled and assigned to three groups based on the diagnosed pathology: (i) no shoulder pathology, (ii) chronic rotator cuff tendinopathy, and (iii) degenerative RCTs. In total, 14 patients were included. Venous blood samples ("liquid biopsies") were collected from each patient and serum levels of 187 miRNAs were determined. Subsequently, the change in expression of nine candidate miRNAs was verified in tendon biopsy samples, collected from patients who underwent arthroscopic shoulder surgery between 2015 and 2018. Overall, we identified several miRNAs to be progressively deregulated in sera from patients with either chronic rotator cuff tendinopathy or degenerative RCTs. Importantly, for the several of these miRNAs candidates repression was also evident in tendon biopsies harvested from patients who were treated for a supraspinatus tendon tear. As similar expression profiles were determined for tendon samples, the newly identified systemic miRNA signature has potential as novel diagnostic or prognostic biomarkers for degenerative rotator cuff pathologies. © 2019 The Authors. Journal of Orthopaedic Research® published by Wiley Periodicals, Inc. on behalf of Orthopaedic Research Society. Inc. J Orthop Res 38:202-211, 2020.
Collapse
Affiliation(s)
- Fabian Plachel
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration, Center SalzburgParacelsus Medical UniversitySalzburgAustria,Center for Musculoskeletal Surgery, Campus VirchowCharité UniversitaetsmedizinBerlinGermany
| | | | - Renate Gehwolf
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration, Center SalzburgParacelsus Medical UniversitySalzburgAustria,Austrian Cluster for Tissue RegenerationViennaAustria
| | - Julia Frank
- Vienna Shoulder & Sports ClinicViennaAustria
| | - Herbert Tempfer
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration, Center SalzburgParacelsus Medical UniversitySalzburgAustria,Austrian Cluster for Tissue RegenerationViennaAustria
| | - Christine Lehner
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration, Center SalzburgParacelsus Medical UniversitySalzburgAustria,Austrian Cluster for Tissue RegenerationViennaAustria
| | - Nadja Weissenbacher
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration, Center SalzburgParacelsus Medical UniversitySalzburgAustria,Austrian Cluster for Tissue RegenerationViennaAustria
| | - Andrea Wagner
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration, Center SalzburgParacelsus Medical UniversitySalzburgAustria,Austrian Cluster for Tissue RegenerationViennaAustria
| | | | - Philipp Moroder
- Center for Musculoskeletal Surgery, Campus VirchowCharité UniversitaetsmedizinBerlinGermany
| | | | - Andreas Traweger
- Institute of Tendon and Bone Regeneration, Spinal Cord Injury and Tissue Regeneration, Center SalzburgParacelsus Medical UniversitySalzburgAustria,Austrian Cluster for Tissue RegenerationViennaAustria
| |
Collapse
|
25
|
Gareev IF, Novikova LB, Beylerli OA. Application of microRNA in the therapy of ischemic stroke. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2019. [DOI: 10.15829/1728-8800-2019-5-66-73] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|
26
|
Xue H, Tu Y, Ma T, Wen T, Yang T, Xue L, Cai M, Wang F, Guan M. miR-93-5p attenuates IL-1β-induced chondrocyte apoptosis and cartilage degradation in osteoarthritis partially by targeting TCF4. Bone 2019; 123:129-136. [PMID: 30930294 DOI: 10.1016/j.bone.2019.03.035] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Accepted: 03/26/2019] [Indexed: 12/15/2022]
Abstract
MicroRNAs (miRNAs, miRs) are frequently dysregulated in osteoarthritis (OA), but the role of specific miRNAs in OA remains unclear. In this study, we found that miR-93-5p is underexpressed in human and rat OA-affected cartilage (compared with normal cartilage) as well as in IL-1β-treated chondrocytes. Overexpression of miR-93-5p promoted chondrocyte viability, suppressed chondrocyte apoptosis, and maintained the balance between anabolic and catabolic factors of the extracellular matrix in vitro. Similarly, injection of a miR-93-5p-expressing lentivirus alleviated the destruction of articular cartilage in a rat model of OA (anterior cruciate ligament transection). Furthermore, TCF4 was identified as a direct target gene of miR-93-5p. miR-93-5p directly targeted the 3' untranslated region (3'-UTR) of TCF4 mRNA and repressed TCF4 expression. Overexpression of TCF4 attenuated the effects of miR-93-5p on chondrocyte apoptosis and functions. Finally, analyses of miR-93-5p and TCF4 in OA-affected cartilage tissues revealed that miR-93-5p expression inversely correlated with TCF4 expression. Altogether, these findings indicate that miR-93-5p slows OA progression partially by suppressing TCF4 expression, and this phenomenon may provide novel insights into the function of miRNA in OA.
Collapse
Affiliation(s)
- Huaming Xue
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| | - Yihui Tu
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China.
| | - Tong Ma
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| | - Tao Wen
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| | - Tao Yang
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| | - Long Xue
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| | - Minwei Cai
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| | - Fangxing Wang
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| | - Mengying Guan
- Department of Orthopaedics, Yangpu District Central Hospital affiliated to Tongji University School of Medicine, Shanghai 200090, China
| |
Collapse
|
27
|
Guo Y, Hong W, Wang X, Zhang P, Körner H, Tu J, Wei W. MicroRNAs in Microglia: How do MicroRNAs Affect Activation, Inflammation, Polarization of Microglia and Mediate the Interaction Between Microglia and Glioma? Front Mol Neurosci 2019; 12:125. [PMID: 31133802 PMCID: PMC6522842 DOI: 10.3389/fnmol.2019.00125] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 04/26/2019] [Indexed: 12/31/2022] Open
Abstract
The essential roles of microglia in maintaining homeostasis in the healthy brain and contributing to neuropathology are well documented. Emerging evidence suggests that epigenetic modulation regulates microglial behavior in both physiological and pathological conditions. MicroRNAs (miRNAs) are short, non-coding epigenetic regulators that repress target gene expression mostly via binding to 3'-untranslated region (3'-UTR) of mRNA in a Dicer-dependent manner. Dysregulation of certain miRNAs can contribute to microglial hyper-activation, persistent neuroinflammation, and abnormal macrophage polarization in the brain. These abnormal conditions can support the pathogenesis of neurological disorders such as glioma, Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), stroke, ischemia, and spinal cord injury (SCI). However, the roles of miRNAs in microglia in health and neurological disease have not been systematically summarized. This review will first report the role of Dicer, a key endoribonulease that is responsible for most miRNA biogenesis in microglia. Second, we will focus on recent research about the function of miRNAs in activation, inflammation and polarization of microglia, respectively. In addition, potential crosstalk between microglia and glioma cells via miRNAs will be discussed in this part. Finally, the role of two essential miRNAs, miR-124, and miR-155, in microglia will be highlighted.
Collapse
Affiliation(s)
- Yawei Guo
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wenming Hong
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
- Department of Neurosurgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xinming Wang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Pengying Zhang
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Heinrich Körner
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Jiajie Tu
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| | - Wei Wei
- Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Ministry of Education, Hefei, China
| |
Collapse
|
28
|
Forouzanfar F, Shojapour M, Asgharzade S, Amini E. Causes and Consequences of MicroRNA Dysregulation Following Cerebral Ischemia-Reperfusion Injury. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 18:212-221. [DOI: 10.2174/1871527318666190204104629] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 12/31/2018] [Accepted: 01/25/2019] [Indexed: 12/31/2022]
Abstract
Stroke continues to be a major cause of death and disability worldwide. In this respect, the
most important mechanisms underlying stroke pathophysiology are inflammatory pathways, oxidative
stress, as well as apoptosis. Accordingly, miRNAs are considered as non-coding endogenous RNA
molecules interacting with their target mRNAs to inhibit mRNA translation or reduce its transcription.
Studies in this domain have similarly shown that miRNAs are strongly associated with coronary artery
disease and correspondingly contributed to the brain ischemia molecular processes. To retrieve articles
related to the study subject, i.e. the role of miRNAs involved in inflammatory pathways, oxidative
stress, and apoptosis in stroke from the databases of Web of Science, PubMed (NLM), Open Access
Journals, LISTA (EBSCO), and Google Scholar; keywords including cerebral ischemia, microRNA
(miRNA), inflammatory pathway, oxidative stress, along with apoptosis were used. It was consequently
inferred that, miRNAs could be employed as potential biomarkers for diagnosis and prognosis, as
well as therapeutic goals of cerebral ischemia.
Collapse
Affiliation(s)
- Fatemeh Forouzanfar
- Medical Toxicology Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mana Shojapour
- Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Samira Asgharzade
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Amini
- UKM Medical Centre [HUKM], Department of Medicine, Faculty of Medicine, Malaysia
| |
Collapse
|
29
|
Ding Y, Wang L, Zhao Q, Wu Z, Kong L. MicroRNA‑93 inhibits chondrocyte apoptosis and inflammation in osteoarthritis by targeting the TLR4/NF‑κB signaling pathway. Int J Mol Med 2018; 43:779-790. [PMID: 30569118 PMCID: PMC6317687 DOI: 10.3892/ijmm.2018.4033] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 12/07/2018] [Indexed: 01/16/2023] Open
Abstract
Osteoarthritis (OA) is a serious disease of the articular cartilage, and inflammation has been implicated in its pathogenesis. Previously, microRNAs (miRNAs) have been proposed as novel regulators of inflammation, however, the functional role of microRNAs in regulating inflammation in OA remains to be fully elucidated. The aim of the present study was to investigate the roles of miRNAs in OA inflammation and the underlying molecular mechanism. Firstly, the miRNA expression patterns were analyzed in the articular cartilage tissues from experimental OA mice using an miRNA microarray. miRNA (miR)-93 was identified with particular interest due to its reported effects on apoptosis and inflammation suppression. Subsequently, the expression of miR-93 was further validated in the articular cartilage tissues of OA mice and lipopolysaccharide (LPS)-stimulated primary chondrocytes. Using this LPS-induced chondrocyte injury model, the overexpression of miR-93 enhanced cell viability, improved cell apoptosis and attenuated the inflammatory response, as reflected by reductions in pro-inflammatory cytokines, including tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6. In addition, Toll-like receptor 4 (TLR4), an important regulator of the nuclear factor-κB (NF-κB) signaling pathway, was identified as a direct target of miR-93 in chondrocytes. Furthermore, the restoration of TLR4 markedly abrogated the inhibitory effects of miR-93 on the chondrocyte apoptosis and inflammation induced by LPS. In addition, the overexpression of miR-93 by agomir-miR-93 significantly inhibited the levels of pro-inflammatory cytokines and cell apoptosis, whereas antagomir-93 exacerbated apoptosis and inflammation in vivo. Taken together, the results of the study suggested that miR-93 may be a promising therapeutic target for the treatment of human OA.
Collapse
Affiliation(s)
- Yanjie Ding
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Laifang Wang
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Qing Zhao
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Zhenzhen Wu
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| | - Lingli Kong
- Department of Rheumatology and Immunology, Huaihe Hospital of Henan University, Kaifeng, Henan 475000, P.R. China
| |
Collapse
|
30
|
Rogobete AF, Sandesc D, Bedreag OH, Papurica M, Popovici SE, Bratu T, Popoiu CM, Nitu R, Dragomir T, AAbed HIM, Ivan MV. MicroRNA Expression is Associated with Sepsis Disorders in Critically Ill Polytrauma Patients. Cells 2018; 7:E271. [PMID: 30551680 PMCID: PMC6316368 DOI: 10.3390/cells7120271] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/06/2018] [Accepted: 12/06/2018] [Indexed: 12/16/2022] Open
Abstract
A critically ill polytrauma patient is one of the most complex cases to be admitted to the intensive care unit, due to both the primary traumatic complications and the secondary post-traumatic interactions. From a molecular, genetic, and epigenetic point of view, numerous biochemical interactions are responsible for the deterioration of the clinical status of a patient, and increased mortality rates. From a molecular viewpoint, microRNAs are one of the most complex macromolecular systems due to the numerous modular reactions and interactions that they are involved in. Regarding the expression and activity of microRNAs in sepsis, their usefulness has reached new levels of significance. MicroRNAs can be used both as an early biomarker for sepsis, and as a therapeutic target because of their ability to block the complex reactions involved in the initiation, maintenance, and augmentation of the clinical status.
Collapse
Affiliation(s)
- Alexandru Florin Rogobete
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Dorel Sandesc
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Ovidiu Horea Bedreag
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Marius Papurica
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
- Clinic of Anesthesia and Intensive Care, Emergency County Hospital "Pius Brinzeu", 300723 Timisoara, Romania.
| | - Sonia Elena Popovici
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Tiberiu Bratu
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Calin Marius Popoiu
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Razvan Nitu
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Tiberiu Dragomir
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Hazzaa I M AAbed
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| | - Mihaela Viviana Ivan
- Faculty of Medicine, "Victor Babes" University of Medicine and Pharmacy, 300041 Timisoara, Romania.
| |
Collapse
|
31
|
Yang Q, Zhou J. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia 2018; 67:1017-1035. [DOI: 10.1002/glia.23571] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Qiao‐qiao Yang
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
| | - Jia‐wei Zhou
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology Chinese Academy of Sciences Shanghai China
- University of Chinese Academy of Sciences Shanghai 200031 China
| |
Collapse
|
32
|
Cheray M, Joseph B. Epigenetics Control Microglia Plasticity. Front Cell Neurosci 2018; 12:243. [PMID: 30123114 PMCID: PMC6085560 DOI: 10.3389/fncel.2018.00243] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 07/18/2018] [Indexed: 01/31/2023] Open
Abstract
Microglia, resident immune cells of the central nervous system, fulfill multiple functions in the brain throughout life. These microglial functions range from participation in innate and adaptive immune responses, involvement in the development of the brain and its homeostasis maintenance, to contribution to degenerative, traumatic, and proliferative diseases; and take place in the developing, the aging, the healthy, or the diseased brain. Thus, an impressive level of cellular plasticity, appears as a requirement for the pleiotropic biological functions of microglia. Epigenetic changes, including histone modifications or DNA methylation as well as microRNA expression, are important modifiers of gene expression, and have been involved in cell phenotype regulation and reprogramming and are therefore part of the mechanisms regulating cellular plasticity. Here, we review and discuss the epigenetic mechanisms, which are emerging as contributors to this microglial cellular plasticity and thereby can constitute interesting targets to modulate microglia associated brain diseases, including developmental diseases, neurodegenerative diseases as well as cancer.
Collapse
Affiliation(s)
- Mathilde Cheray
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| | - Bertrand Joseph
- Toxicology Unit, Institute of Environmental Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
33
|
Abstract
Central nervous system (CNS) injuries, such as stroke, traumatic brain injury (TBI) and spinal cord injury (SCI), are important causes of death and long-term disability worldwide. MicroRNA (miRNA), small non-coding RNA molecules that negatively regulate gene expression, can serve as diagnostic biomarkers and are emerging as novel therapeutic targets for CNS injuries. MiRNA-based therapeutics include miRNA mimics and inhibitors (antagomiRs) to respectively decrease and increase the expression of target genes. In this review, we summarize current miRNA-based therapeutic applications in stroke, TBI and SCI. Administration methods, time windows and dosage for effective delivery of miRNA-based drugs into CNS are discussed. The underlying mechanisms of miRNA-based therapeutics are reviewed including oxidative stress, inflammation, apoptosis, blood-brain barrier protection, angiogenesis and neurogenesis. Pharmacological agents that protect against CNS injuries by targeting specific miRNAs are presented along with the challenges and therapeutic potential of miRNA-based therapies.
Collapse
Affiliation(s)
- Ping Sun
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Da Zhi Liu
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, CA, USA
| | - Glen C Jickling
- Department of Neurology, University of Alberta, Edmonton, Alberta, Canada
| | - Frank R Sharp
- Department of Neurology and the M.I.N.D. Institute, University of California at Davis, Sacramento, CA, USA
| | - Ke-Jie Yin
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Ke-Jie Yin, Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery, University of Pittsburgh School of Medicine, 200 Lothrop Street, BST S514, Pittsburgh, PA 15213, USA. Da Zhi Liu, Department of Neurology, University of California at Davis, Sacramento, CA 95817, USA.
| |
Collapse
|
34
|
Xie YL, Zhang B, Jing L. MiR-125b blocks Bax/Cytochrome C/Caspase-3 apoptotic signaling pathway in rat models of cerebral ischemia-reperfusion injury by targeting p53. Neurol Res 2018; 40:828-837. [PMID: 29956588 DOI: 10.1080/01616412.2018.1488654] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To explore the potential effect of miR-125b on p53-mediated regulation of Bax/Cytochrome C/Caspase-3 apoptotic signaling pathway in rats with cerebral ischemia-reperfusion (CIR) injury. METHODS Sprague-Dawley (SD) rats were used to conduct CIR injury and injected with miR-125b mimic/inhibitor or p53 inhibitor (Pifithrin-α, PFT-α). Dual-luciferase reporter gene assay was used to analyze the targeting relationship between miR-125b and p53. Longa scoring and Triphenyl tetrazolinm chloride (TTC) staining were used to test the neurologic function and determine infarct size, respectively. Hematoxylin-eosin (HE) and Nissl's stainings were conducted to observe the morphology of cortical neurons. Neuronal nuclei (NeuN) expression was detected by immunohistochemical staining. QRT-PCR was performed to detect the expressions of miR-125b and p53. TUNEL staining and Western blotting was used to determine neuronal apoptosis and expressions of Bax/Cytochrome C/Caspase-3 signaling pathway-related proteins, respectively. RESULTS Our results showed that miR-125b could directly target p53. As observed, overexpression of miR-125b could obviously reduce the neurological score, infarct size, and brain water content after CIR in rats, which also improved the morphology of cortical neurons, increased the number of neurons, reduced neuronal apoptosis, and inhibited the expressions of Bax/Cytochrome C/Caspase-3 pathway. Moreover,the similar results were observed in rats with CIR after injected with PFT-α. But no significant differences in each index were found in CIR group and CIR + anti-miR-125b + PFT-α group. CONCLUSION MiR-125b exerts protective effects on CIR injury through inhibition of Bax/Cytochrome C/Caspase-3signaling pathway via targeting p53, which is likely to be a promising treatment for CIR. ABBREVIATIONS 3'-UTR: 3-untranslated region; CIR: cerebral ischemia-reperfusion; CIS: cerebral ischemic stroke; PFT-α: Pifithrin-α; PVDF: polyvinylidene fluoride; SD: Sprague-Dawley; TBST: tris buffered saline with tween. TTC staining: Triphenyl tetrazolinm chloride staining; TUNEL: Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling.
Collapse
Affiliation(s)
- Yun-Liang Xie
- a Medical Department , The Affiliated Hospital of Bei Hua University , Jilin , China
| | - Bo Zhang
- b Health Care Department , The Affiliated Hospital of Bei Hua University , Jilin , China
| | - Ling Jing
- c College of Pharmacy , Jilin University , Changchun , China
| |
Collapse
|
35
|
The Role of Circular RNAs in Cerebral Ischemic Diseases: Ischemic Stroke and Cerebral Ischemia/Reperfusion Injury. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1087:309-325. [PMID: 30259377 DOI: 10.1007/978-981-13-1426-1_25] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Cerebral ischemic diseases including ischemic stroke and cerebral ischemia reperfusion injury can result in serious dysfunction of the brain, which leads to extremely high mortality and disability. There are no effective therapeutics for cerebral ischemic diseases to date. Circular RNAs are a kind of newly investigated noncoding RNAs. It is reported that circular RNAs are enriched in multiple organs, especially abundant in the brain, which indicates that circular RNAs may be involved in cerebral physiological and pathological processes. In this chapter, we will firstly review the pathophysiology, underlying mechanisms, and current treatments of cerebral ischemic diseases including ischemic stroke and cerebral ischemia/reperfusion injury. Secondly, the characteristics and function of circular RNAs will be outlined, and then we are going to introduce the roles circular RNAs play in human diseases. Finally, we will summarize the function of circular RNAs in cerebral ischemic diseases.
Collapse
|