1
|
Liu J, Tan G, Wang S, Tong B, Wu Y, Zhang L, Jiang B. Artesunate induces HO-1-mediated cell cycle arrest and senescence to protect against ocular fibrosis. Int Immunopharmacol 2024; 141:112882. [PMID: 39151383 DOI: 10.1016/j.intimp.2024.112882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/04/2024] [Accepted: 08/04/2024] [Indexed: 08/19/2024]
Abstract
Recent research found artesunate could inhibit ocular fibrosis; however, the underlying mechanisms are not fully known. Since the ocular fibroblast is the main effector cell in fibrosis, we hypothesized that artesunate may exert its protective effects by inhibiting the fibroblasts proliferation. TGF-β1-induced ocular fibroblasts and glaucoma filtration surgery (GFS)-treated rabbits were used as ocular fibrotic models. Firstly, we analyzed fibrosis levels by assessing the expression of fibrotic marker proteins, and used Ki67 immunofluorescence, EdU staining, flow cytometry to determine cell cycle status, and SA-β-gal staining to assess cellular senescence levels. Then to predict target genes and pathways of artesunate, we analyzed the differentially expressed genes and enriched pathways through RNA-seq. Western blot and immunohistochemistry were used to detect the pathway-related proteins. Additionally, we validated the dependence of artesunate's effects on HO-1 expression through HO-1 siRNA. Moreover, DCFDA and MitoSOX fluorescence staining were used to examine ROS level. We found artesunate significantly inhibits the expression of fibrosis-related proteins, induces cell cycle arrest and cellular senescence. Knocking down HO-1 in fibroblasts with siRNA reverses these regulatory effects of artesunate. Mechanistic studies show that artesunate significantly inhibits the activation of the Cyclin D1/CDK4-pRB pathway, induces an increase in cellular and mitochondrial ROS levels and activates the Nrf2/HO-1 pathway. In conclusion, the present study identifies that artesunate induces HO-1 expression through ROS to activate the antioxidant Nrf2/HO-1 pathway, subsequently inhibits the cell cycle regulation pathway Cyclin D1/CDK4-pRB in an HO-1-dependent way, induces cell cycle arrest and senescence, and thereby resists periorbital fibrosis.
Collapse
Affiliation(s)
- Jingyuan Liu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Guangshuang Tan
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Shutong Wang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Boding Tong
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Ying Wu
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China
| | - Lusi Zhang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China.
| | - Bing Jiang
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410000, Hunan, China; Hunan Clinical Research Center of Ophthalmic Disease, Changsha, Hunan 410011, China.
| |
Collapse
|
2
|
Li C, Wu J, Dong Q, Ma J, Gao H, Liu G, Chen Y, Ning J, Lv X, Zhang M, Zhong H, Zheng T, Liu Y, Peng Y, Qu Y, Gao X, Shi H, Sun C, Hui Y. The crosstalk between oxidative stress and DNA damage induces neural stem cell senescence by HO-1/PARP1 non-canonical pathway. Free Radic Biol Med 2024; 223:443-457. [PMID: 39047850 DOI: 10.1016/j.freeradbiomed.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 07/19/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Neural stem cells play a crucial role in maintaining brain homeostasis. Neural stem cells senescence can lead to the decline of nerve repair and regeneration, causing brain aging and neurodegenerative diseases. However, the mechanism underlying neural stem cells senescence remains poorly understood. In this study, we report a novel HO-1/PARP1 non-canonical pathway highlighting how oxidative stress triggers the DNA damage response, ultimately leading to premature cellular senescence in neural stem cells. HO-1 acts as a sensor for oxidative stress, while PARP1 functions as a sensor for DNA damage. The simultaneous expression and molecular interaction of these two sensors can initiate a crosstalk of oxidative stress and DNA damage response processes, leading to the vicious cycle. The persistent activation of this pathway contributes to the senescence of neural stem cells, which in turn plays a crucial role in the progression of neurodegenerative diseases. Consequently, targeting this novel signaling pathway holds promise for the development of innovative therapeutic strategies and targets aimed at mitigating neural stem cells senescence-related disorders.
Collapse
Affiliation(s)
- Cheng Li
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiajia Wu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Qi Dong
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiajia Ma
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Huiqun Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Guiyan Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - You Chen
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Jiaqi Ning
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Xuebing Lv
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Mingyang Zhang
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Haojie Zhong
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Tianhu Zheng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yuanli Liu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yahui Peng
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Yilin Qu
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China
| | - Xu Gao
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China; Basic Medical Institute of Heilongjiang Medical Science Academy, PR China; Translational Medicine Center of Northern China, PR China
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, PR China.
| | - Chongran Sun
- Department of Neurosurgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, PR China.
| | - Yang Hui
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, PR China; Basic Medical Institute of Heilongjiang Medical Science Academy, PR China; Translational Medicine Center of Northern China, PR China.
| |
Collapse
|
3
|
Shi YS, Chen JC, Lin BH, Wang RN, Zhao J, Li S, Zhang Y, Zhang XF. Pteris laeta Wall. and Its New Phytochemical, Pterosinsade A, Promote Hippocampal Neurogenesis via Activating the Wnt Signaling Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:4586-4598. [PMID: 36892329 DOI: 10.1021/acs.jafc.2c08493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Pteris laeta Wall., as a traditional tea, is popular in Southwest China, but its role in preventing cognitive impairment is unclear. In this study, Pteris laeta Wall. extracts (PW) and its active compounds were evaluated for preventive effects on Alzheimer's disease (AD) in vivo and in vitro. The results showed that PW diminished oxidative stress damage and apoptosis of Aβ-induced HT22 cells and also rescued cognitive deficits, and ameliorated pathological injury and inflammatory response in APP/PS1 mice. Besides, a new pterosin sesquiterpene, named pterosinsade A (PA), and nine known compounds were discovered from the EtOAc extract that possessed the best neuroprotective activity. PA reduced apoptosis of APP-overexpressing neural stem cells and promoted their proliferation and neuronal differentiation. Meanwhile, PW and PA promoted hippocampal neurogenesis, which proved to be associated with activating the Wnt signaling pathway. These findings suggest that PW and PA are candidates for AD prevention.
Collapse
Affiliation(s)
- Yu-Sheng Shi
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, China
- Academy of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Ji-Cong Chen
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Bi-Hui Lin
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510920, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Ruo-Nan Wang
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, China
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116600, China
| | - Jie Zhao
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, China
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116600, China
| | - Sheng Li
- National-Local Joint Engineering Research Center for Drug-Research and Development (R&D) of Neurodegenerative Diseases, Dalian Medical University, Dalian 116044, China
- College of Basic Medical Sciences, Dalian Medical University, Dalian 116600, China
| | - Yan Zhang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510920, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
- Chiba University, Matsudo 2718501, Japan
| | - Xu-Fu Zhang
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou 510920, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
4
|
Zhou Y, Wang X, Liu Y, Gu Y, Gu R, Zhang G, Lin Q. Mechanisms of abnormal adult hippocampal neurogenesis in Alzheimer's disease. Front Neurosci 2023; 17:1125376. [PMID: 36875663 PMCID: PMC9975352 DOI: 10.3389/fnins.2023.1125376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 01/31/2023] [Indexed: 02/17/2023] Open
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system, the most common type of dementia in old age, which causes progressive loss of cognitive functions such as thoughts, memory, reasoning, behavioral abilities and social skills, affecting the daily life of patients. The dentate gyrus of the hippocampus is a key area for learning and memory functions, and an important site of adult hippocampal neurogenesis (AHN) in normal mammals. AHN mainly consists of the proliferation, differentiation, survival and maturation of newborn neurons and occurs throughout adulthood, but the level of AHN decreases with age. In AD, the AHN will be affected to different degrees at different times, and its exact molecular mechanisms are increasingly elucidated. In this review, we summarize the changes of AHN in AD and its alteration mechanism, which will help lay the foundation for further research on the pathogenesis and diagnostic and therapeutic approaches of AD.
Collapse
Affiliation(s)
- Yujuan Zhou
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Xu Wang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Yingying Liu
- Department of Physiology and Pathophysiology, Health Science Center, Peking University, Beijing, China
| | - Yulu Gu
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
| | - Renjun Gu
- School of Chinese Medicine and School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Geng Zhang
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
- Laboratory of Clinical Applied Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Qing Lin
- Key Laboratory of Brain Aging and Neurodegenerative Diseases, Fujian Medical University, Fuzhou, China
- Laboratory of Clinical Applied Anatomy, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
5
|
Kasica NP, Zhou X, Jester HM, Holland CE, Ryazanov AG, Forshaw TE, Furdui CM, Ma T. Homozygous knockout of eEF2K alleviates cognitive deficits in APP/PS1 Alzheimer’s disease model mice independent of brain amyloid β pathology. Front Aging Neurosci 2022; 14:959326. [PMID: 36158543 PMCID: PMC9500344 DOI: 10.3389/fnagi.2022.959326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/23/2022] [Indexed: 11/25/2022] Open
Abstract
Maintenance of memory and synaptic plasticity depends on de novo protein synthesis, and accumulating evidence implicates a role of dysregulated mRNA translation in cognitive impairments associated with Alzheimer’s disease (AD). Accumulating evidence demonstrates hyper-phosphorylation of translation factor eukaryotic elongation factor 2 (eEF2) in the hippocampi of human AD patients as well as transgenic AD model mice. Phosphorylation of eEF2 (at the Thr 56 site) by its only known kinase, eEF2K, leads to inhibition of general protein synthesis. A recent study suggests that amyloid β (Aβ)-induced neurotoxicity could be associated with an interaction between eEF2 phosphorylation and the transcription factor nuclear erythroid 2-related factor (NRF2)-mediated antioxidant response. In this brief communication, we report that global homozygous knockout of the eEF2K gene alleviates deficits of long-term recognition and spatial learning in a mouse model of AD (APP/PS1). Moreover, eEF2K knockout does not alter brain Aβ pathology in APP/PS1 mice. The hippocampal NRF2 antioxidant response in the APP/PS1 mice, measured by expression levels of nicotinamide adenine dinucleotide plus hydrogen (NADPH) quinone oxidoreductase 1 (NQO1) and heme oxygenase-1 (HO-1), is ameliorated by suppression of eEF2K signaling. Together, the findings may contribute to our understanding of the molecular mechanisms underlying AD pathogenesis, indicating that suppression of eEF2K activity could be a beneficial therapeutic option for this devastating neurodegenerative disease.
Collapse
Affiliation(s)
- Nicole P. Kasica
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Xueyan Zhou
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Hannah M. Jester
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Caroline E. Holland
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Alexey G. Ryazanov
- Department of Pharmacology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, United States
| | - Tom E. Forshaw
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Cristina M. Furdui
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Tao Ma
- Department of Internal Medicine, Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- Department of Neurobiology and Anatomy, Wake Forest University School of Medicine, Winston-Salem, NC, United States
- *Correspondence: Tao Ma,
| |
Collapse
|
6
|
Characteristics, Classification, and Application of Stem Cells Derived from Human Teeth. Stem Cells Int 2021; 2021:8886854. [PMID: 34194509 PMCID: PMC8184333 DOI: 10.1155/2021/8886854] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 02/12/2021] [Accepted: 05/07/2021] [Indexed: 12/31/2022] Open
Abstract
Since mesenchymal stem cells derived from human teeth are characterized as having the properties of excellent proliferation, multilineage differentiation, and immune regulation. Dental stem cells exhibit fibroblast-like microscopic appearance and express mesenchymal markers, embryonic markers, and vascular markers but do not express hematopoietic markers. Dental stem cells are a mixed population with different sensitive markers, characteristics, and therapeutic effects. Single or combined surface markers are not only helpful for understanding the subpopulation of mixed stem cell populations according to cell function but also for improving the stable treatment effect of dental stem cells. Focusing on the discovery and characterization of stem cells isolated from human teeth over the past 20 years, this review outlines the effect of marker sorting on cell proliferation and differentiation ability and the assessment of the clinical application potential. Classified dental stem cells from markers and functional molecules can solve the problem of heterogeneity and ensure the efficacy of cell therapy strategies including dentistry, neurologic diseases, bone repair, and tissue engineering.
Collapse
|
7
|
Si Z, Wang X. Stem Cell Therapies in Alzheimer's Disease: Applications for Disease Modeling. J Pharmacol Exp Ther 2021; 377:207-217. [PMID: 33558427 DOI: 10.1124/jpet.120.000324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/03/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease with complex pathologic and biologic characteristics. Extracellular β-amyloid deposits, such as senile plaques, and intracellular aggregation of hyperphosphorylated tau, such as neurofibrillary tangles, remain the main neuropathological criteria for the diagnosis of AD. There is currently no effective treatment of the disease, and many clinical trials have failed to prove any benefits of new therapeutics. More recently, there has been increasing interest in harnessing the potential of stem cell technologies for drug discovery, disease modeling, and cell therapies, which have been used to study an array of human conditions, including AD. The recently developed and optimized induced pluripotent stem cell (iPSC) technology is a critical platform for screening anti-AD drugs and understanding mutations that modify AD. Neural stem cell (NSC) transplantation has been investigated as a new therapeutic approach to treat neurodegenerative diseases. Mesenchymal stem cells (MSCs) also exhibit considerable potential to treat neurodegenerative diseases by secreting growth factors and exosomes, attenuating neuroinflammation. This review highlights recent progress in stem cell research and the translational applications and challenges of iPSCs, NSCs, and MSCs as treatment strategies for AD. Even though these treatments are still in relative infancy, these developing stem cell technologies hold considerable promise to combat AD and other neurodegenerative disorders. SIGNIFICANCE STATEMENT: Alzheimer's disease (AD) is a neurodegenerative disease that results in learning and memory defects. Although some drugs have been approved for AD treatment, fewer than 20% of patients with AD benefit from these drugs. Therapies based on stem cells, including induced pluripotent stem cells, neural stem cells, and mesenchymal stem cells, provide promising therapeutic strategies for AD.
Collapse
Affiliation(s)
- Zizhen Si
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| | - Xidi Wang
- Department of Physiology and Pharmacology, School of Medicine, Ningbo University, Ningbo, China (Z.S.) and Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, China (X.W.)
| |
Collapse
|
8
|
Si Z, Zhou S, Shen Z, Luan F, Yan J. lncRNA HAND2-AS1 is downregulated in osteoarthritis and regulates IL-6 expression in chondrocytes. J Orthop Surg Res 2021; 16:68. [PMID: 33468183 PMCID: PMC7816418 DOI: 10.1186/s13018-021-02216-9] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Osteoarthritis (OA) is a leading cause of disability. The incidence of OA is progressively rising due to the diminishing levels of physical activity and ever-expanding aging population. However, the mainstay for OA treatment only can improve symptoms without delay the progression of this severe disease. This study aimed to explore the biological role and clinical function of lncRNA HAND2-AS1 in OA. METHODS Blood samples and synovial fluid were collected from OA patients and normal subjects. HAND2-AS1 expression was detected by qRT-PCR and IL-6 expression was detected by ELISA. The plasma levels of HAND2-AS1 were also detected in different ages, stages, and gender of OA patients and controls. Furthermore, the ROC curve was used to analyze whether HAND2-AS1 can distinguish OA patients from normal subjects. Also, Pearson correlation coefficient analysis was used to analyze the correlation between lncRNA HAND2-AS1 and IL-6. In addition, Western blot was used to detect the IL-6 level upon HAND2-AS1 over-expression in chondrocytes and qRT-PCR was used to detect the HAND2-AS1 level after endogenous IL-6 treatment. RESULTS HAND2-AS1 and IL-6 were dysregulated in plasma and synovial fluid of OA patients. The expression of HAND2-AS1 in plasma of OA patients was decreased with aging and progression. Furthermore, HAND2-AS1 downregulation effectively distinguished OA patients from the healthy controls. Over-expression of HAND2-AS1 inhibited IL-6 expression in chondrocytes, while treatment with exogenous IL-6 did not affect HAND2-AS1 expression. CONCLUSION HAND2-AS1 effectively distinguished OA patients from the healthy controls and regulates IL-6 expression in human chondrocytes. TRIAL REGISTRATION ChiCTR, ChiCTR2000038635 . Registered 11 February 2019.
Collapse
Affiliation(s)
- Zhenxing Si
- Orthopedic Department, Second Affiliated Hospital of Harbin Medical University, No. 148 Baojian Road, Nangang District, Harbin, Heilongjiang, China
| | - Shifeng Zhou
- Surgical Emergency, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zilong Shen
- Orthopedic Department, Second Affiliated Hospital of Harbin Medical University, No. 148 Baojian Road, Nangang District, Harbin, Heilongjiang, China
| | - Feiyu Luan
- Surgical Emergency, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Orthopedic Department, Second Affiliated Hospital of Harbin Medical University, No. 148 Baojian Road, Nangang District, Harbin, Heilongjiang, China.
| |
Collapse
|