1
|
Li X, Yuan F, Xiong Y, Tang Y, Li Z, Ai J, Miao J, Ye W, Zhou S, Wu Q, Wang X, Xu D, Li J, Huang J, Chen Q, Shen W, Liu Y, Hou FF, Zhou L. FAM3A plays a key role in protecting against tubular cell pyroptosis and acute kidney injury. Redox Biol 2024; 74:103225. [PMID: 38875957 PMCID: PMC11226986 DOI: 10.1016/j.redox.2024.103225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 05/21/2024] [Accepted: 06/04/2024] [Indexed: 06/16/2024] Open
Abstract
Acute kidney injury (AKI) is in high prevalence worldwide but with no therapeutic strategies. Programmed cell death in tubular epithelial cells has been reported to accelerate a variety of AKI, but the major pathways and underlying mechanisms are not defined. Herein, we identified that pyroptosis was responsible for AKI progression and related to ATP depletion in renal tubular cells. We found that FAM3A, a mitochondrial protein that assists ATP synthesis, was decreased and negatively correlated with tubular cell injury and pyroptosis in both mice and patients with AKI. Knockout of FAM3A worsened kidney function decline, increased macrophage and neutrophil cell infiltration, and facilitated tubular cell pyroptosis in ischemia/reperfusion injury model. Conversely, FAM3A overexpression alleviated tubular cell pyroptosis, and inhibited kidney injury in ischemic AKI. Mechanistically, FAM3A promoted PI3K/AKT/NRF2 signaling, thus blocking mitochondrial reactive oxygen species (mt-ROS) accumulation. NLRP3 inflammasome sensed the overload of mt-ROS and then activated Caspase-1, which cleaved GSDMD, pro-IL-1β, and pro-IL-18 into their mature forms to mediate pyroptosis. Of interest, NRF2 activator alleviated the pro-pyroptotic effects of FAM3A depletion, whereas the deletion of NRF2 blocked the anti-pyroptotic function of FAM3A. Thus, our study provides new mechanisms for AKI progression and demonstrates that FAM3A is a potential therapeutic target for treating AKI.
Collapse
Affiliation(s)
- Xiaolong Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Feifei Yuan
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yabing Xiong
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ying Tang
- Department of Nephrology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhiru Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jun Ai
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinhua Miao
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenting Ye
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shan Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qinyu Wu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxu Wang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Dan Xu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiemei Li
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jiewu Huang
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiurong Chen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Weiwei Shen
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Youhua Liu
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Fan Hou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Lili Zhou
- State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Guangdong Provincial Clinical Research Center for Kidney Disease, Guangdong Provincial Key Laboratory of Nephrology, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
2
|
Zhang Z, Guo R, Cai C, Guo P. Down-regulation of long noncoding RNA HOXA11-AS nullifies the impact of microRNA-506-3p on chondrocytes proliferation and apoptosis in osteoarthritis. Clinics (Sao Paulo) 2024; 79:100393. [PMID: 38815540 PMCID: PMC11177079 DOI: 10.1016/j.clinsp.2024.100393] [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: 09/13/2023] [Revised: 04/17/2024] [Accepted: 05/13/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVES This study was directed towards exploring the impacts of lncRNA HOXA11-AS-mediated microRNA (miR)-506-3p on chondrocytes proliferation and apoptosis in osteoarthritis (OA). METHODS The articular cartilages were provided by OA patients who received total knee arthroplasty, and Human Chondrocyte (HC)-OA (HCOA) was also attained. The miR-506-3p and HOXA11-AS expressions in articular cartilages from OA patients and HCOA cells were analyzed via qPCR. After gain- and loss-of-function assays in HCOA cells, MTT assay and flow cytometry (FC) were used for assessing cell viability and apoptosis, accordingly. The levels of PIK3CA, AKT, and mTOR as well as AKT and mTOR phosphorylation levels assessed using western blotting (WB). The targeting correlation of HOXA11-AS and miR-506-3p as well as miR-506-3p and PIK3CA was assessed through Dual-Luciferase Reporter gene Assay (DLRA). RESULT The articular cartilages from OA patients and Human Chondrocyte (HC)-OA (HCOA) cells showed increased HOXA11-AS and decreased miR-506-3p. Mechanistically, HOXA11-AS was capable of binding to miR-506-3p to increase PIK3CA, the target gene of miR-506-3p. miR-506-3p suppression facilitated HCOA cell proliferation and reduced their apoptosis, which was nullified by further silencing HOXA11-AS or silencing PIK3CA. The down-regulation of HOXA11-AS disrupted the PI3K/AKT/mTOR pathway, which was counteracted by further miR-506-3p inhibition. CONCLUSION The silencing of HOXA11-AS might block the PI3K/AKT/mTOR pathway through miR-506-3p up-regulation, thereby restricting HCOA cell proliferation and provoking apoptosis.
Collapse
Affiliation(s)
- Ziyang Zhang
- Department of Gdansk University of Physical Education and Sport, Start for Ph.D. in Sport & Fitness Science, Gdansk, Poland
| | - Renhao Guo
- Department of Olympic Sports Training Theory, National University of Ukraine on Physical Education and Sport, Start for PhD. St. 1, Ukraine
| | - Chengfa Cai
- Department of Exercise Physiology and Biochemistry, Shandong Institute of Sport Science, Shandong, China
| | - Pengcheng Guo
- Department of Key Lab of Aquatic Sports Training Monitoring and Intervention of General Administration of Sport of China, Faculty of Physical Education, Jiangxi Normal University, Jiangxi, China.
| |
Collapse
|
3
|
Zhang FF, Zhang L, Zhao L, Lu Y, Dong X, Liu YQ, Li Y, Guo S, Zheng SY, Xiao Y, Jiang YZ. The circular RNA Rap1b promotes Hoxa5 transcription by recruiting Kat7 and leading to increased Fam3a expression, which inhibits neuronal apoptosis in acute ischemic stroke. Neural Regen Res 2023; 18:2237-2245. [PMID: 37056143 PMCID: PMC10328278 DOI: 10.4103/1673-5374.369115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 10/15/2022] [Accepted: 12/30/2022] [Indexed: 02/17/2023] Open
Abstract
Circular RNAs can regulate the development and progression of ischemic cerebral disease. However, it remains unclear whether they play a role in acute ischemic stroke. To investigate the role of the circular RNA Rap1b (circRap1b) in acute ischemic stroke, in this study we established an in vitro model of acute ischemia and hypoxia by subjecting HT22 cells to oxygen and glucose deprivation and a mouse model of acute ischemia and hypoxia by occluding the right carotid artery. We found that circRap1b expression was remarkably down-regulated in the hippocampal tissue of the mouse model and in the HT22 cell model. In addition, Hoxa5 expression was strongly up-regulated in response to circRap1b overexpression. Hoxa5 expression was low in the hippocampus of a mouse model of acute ischemia and in HT22-AIS cells, and inhibited HT22-AIS cell apoptosis. Importantly, we found that circRap1b promoted Hoxa5 transcription by recruiting the acetyltransferase Kat7 to induce H3K14ac modification in the Hoxa5 promoter region. Hoxa5 regulated neuronal apoptosis by activating transcription of Fam3a, a neuronal apoptosis-related protein. These results suggest that circRap1b regulates Hoxa5 transcription and expression, and subsequently Fam3a expression, ultimately inhibiting cell apoptosis. Lastly, we explored the potential clinical relevance of circRap1b and Hoxa5 in vivo. Taken together, these findings demonstrate the mechanism by which circRap1b inhibits neuronal apoptosis in acute ischemic stroke.
Collapse
Affiliation(s)
- Fang-Fang Zhang
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Liang Zhang
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Lin Zhao
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yu Lu
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Xin Dong
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yan-Qi Liu
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yu Li
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Shuang Guo
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Si-Yuan Zheng
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Ying Xiao
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| | - Yu-Zhu Jiang
- Department of Rehabilitation Medicine, The People’s Hospital of China Medical University (The People’s Hospital of Liaoning Province), Shenyang, Liaoning Province, China
| |
Collapse
|
4
|
Dong QT, Ma DD, Gong Q, Lin ZY, Li ZH, Ye JX, Qin CH, Jin WD, Zhang JX, Zhang ZY. FAM3 family genes are associated with prognostic value of human cancer: a pan-cancer analysis. Sci Rep 2023; 13:15144. [PMID: 37704682 PMCID: PMC10499837 DOI: 10.1038/s41598-023-42060-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] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 09/05/2023] [Indexed: 09/15/2023] Open
Abstract
Family with sequence similarity three member (FAM3) plays a crucial role in the malignant development of various cancers of human. However, there remains doubtful what specific role of FAM3 family genes in pan-cancer. Our study aimed to investigate the role of FAM3 family genes in prognosis, immune subtype, tumor immune microenvironment, stemness score, and anticancer drug sensitivity of pan-cancer. We obtained data from UCSC Xena GDC and CellMiner databases, and used them to study the correlation of the expression, survival, immune subtype, tumor microenvironment, stemness score, and anticancer drug sensitivity between FAM3 family genes with pan-cancer. Furthermore, we investigated the tumor cellular functions and clinical prognostic value FAMC3 in pancreatic cancer (PAAD) using cellular experiments and tissue microarray. Cell Counting Kit-8 (CCK-8), transwell invasion, wound-healing and apoptosis assays were performed to study the effect of FAM3C on SW1990 cells' proliferation, migration, invasion and apoptosis. Immunohistochemical staining was used to study the relationship between FAM3C expression and clinical characteristics of pancreatic cancer patients. The results revealed that FAM3 family genes are significantly differential expression in tumor and adjacent normal tissues in 7 cancers (CHOL, HNSC, KICH, LUAD, LUSC, READ, and STAD). The expression of FAM3 family genes were negatively related with the RNAss, and robust correlated with immune type, tumor immune microenvironment and drug sensitivity. The expression of FAM3 family genes in pan-cancers were significantly different in immune type C1 (wound healing), C2 (IFN-gamma dominant), C3 (inflammatory), C4 (lymphocyte depleted), C5 (immunologically quiet), and C6 (TGF-beta dominant). Meanwhile, overexpression FAM3C promoted SW1990 cells proliferation, migration, invasion and suppressed SW1990 cells apoptosis. While knockdown of FAM3C triggered opposite results. High FAM3C expression was associated with duodenal invasion, differentiation and liver metastasis. In summary, this study provided a new perspective on the potential therapeutic role of FAM3 family genes in pan-cancer. In particular, FAM3C may play an important role in the occurrence and progression of PAAD.
Collapse
Affiliation(s)
- Qing-Tai Dong
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Dan-Dan Ma
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
| | - Qi Gong
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
| | - Zhen-Yu Lin
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhong-Hu Li
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
| | - Jia-Xin Ye
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
| | - Chun-Hui Qin
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
| | - Wei-Dong Jin
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Jian-Xin Zhang
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China.
| | - Zhi-Yong Zhang
- Department of General Surgery, General Hospital of Central Theater Command, Wuhan, 430070, Hubei, China.
| |
Collapse
|
5
|
Yang L, Du B, Zhang S, Wang M. FAM3A mediates the phenotypic switch of human aortic smooth muscle cells stimulated with oxidised low-density lipoprotein by influencing the PI3K-AKT pathway. In Vitro Cell Dev Biol Anim 2023; 59:431-442. [PMID: 37474885 DOI: 10.1007/s11626-023-00775-1] [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: 04/11/2023] [Accepted: 05/26/2023] [Indexed: 07/22/2023]
Abstract
Family with sequence similarity 3 member A (FAM3A) is a multifunctional protein that is related to the pathological process of various disorders. FAM3A is reportedly able to affect the phenotypic change of vascular smooth muscle cells under a hypertensive state. Whether FAM3A mediates the phenotypic switch of vascular smooth muscle cells under an atherosclerotic state remains unaddressed. This work investigated the roles and mechanisms of FAM3A in mediating the phenotypic switch of human aortic smooth muscle cells (HASMCs) stimulated with oxidised low-density lipoprotein (ox-LDL) in vitro. FAM3A expression was elevated in HASMCs following ox-LDL treatment. FAM3A silencing led to a suppressive effect on ox-LDL-provoked proliferation, migration and inflammation of HASMCs, whereas FAM3A overexpression had an opposite effect. Ox-LDL elicited a change in HASMCs from a contractile phenotype to a synthetic phenotype, which was inhibited by FAM3A silencing or enhanced by FAM3A overexpression. Further investigation elucidated that FAM3A silencing repressed and FAM3A overexpression promoted ox-LDL-induced activation of the PI3K-AKT pathway in HASMCs. Reactivation of AKT reversed the suppressive effect of FAM3A silencing on the ox-LDL-induced phenotypic switch of HASMCs. Restraining AKT blocked the promoting effect of FAM3A overexpression on the ox-LDL-induced phenotypic switch of HASMCs. In summary, this work elucidates that FAM3A mediates the ox-LDL-induced phenotypic switch of HASMCs by influencing the PI3K-AKT pathway, indicating a potential role for FAM3A in atherosclerosis.
Collapse
Affiliation(s)
- Lei Yang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an City, Shaanxi Province, 710038, People's Republic of China
| | - Baoshun Du
- Second Department of Neurosurgery, Xinxiang Central Hospital, Xinxiang, Henan Province, 453003, People's Republic of China
| | - Shitao Zhang
- Department of Neurosurgery, Xi'an No. 3 Hospital, The Affiliated Hospital of Northwest University, Xi'an, Shaanxi Province, 710018, People's Republic of China
| | - Maode Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, 277 Yanta West Road, Xi'an City, Shaanxi Province, 710038, People's Republic of China.
| |
Collapse
|
6
|
Wang P, Qian H, Xiao M, Lv J. Role of signal transduction pathways in IL-1β-induced apoptosis: Pathological and therapeutic aspects. Immun Inflamm Dis 2023; 11:e762. [PMID: 36705417 PMCID: PMC9837938 DOI: 10.1002/iid3.762] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/20/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Interleukin-1β (IL-1β) is a pro-inflammatory cytokine mainly produced by monocytes and macrophages with a wide range of biological effects. Evidence has shown that IL-1β plays a vital role in the process of apoptosis; however, the specific mechanisms, by which IL-1β induces apoptosis, vary due to different cellular and experimental conditions. Therefore, this present reviewstudy aimed to systematically review the association between the molecular mechanisms of IL-1β-induced apoptosis in pathological processes and the role of signaling pathways. This article also sought to briefly investigate the potential of signaling pathway-targeted therapy in the prevention and treatment of disease. METHODS This is a literature review article. The present discourse aim is first to scrutinize and assess the available literature on IL-1β and apoptosis. The relevant studies using the keywords of "IL-1β-induced apoptosis" and "signaling pathways" were searched in the databases of PubMed, Scopus, Google Scholar, and Web of Science. Gathered relevant material, and extracted information was then assessed. RESULTS IL-1β can induce apoptosis in various types of cells under different external stimuli via the mitochondrial pathway, death receptor pathway and endoplasmic reticulum pathway, and that the different pathways are often interconnected. The NF-kB signaling pathway, p38MAPK, and JNK signaling pathways mainly play a proapoptotic part, and the ERK1/2 pathway has a bidirectional role in regulating apoptosis, while activation of the PI3K-Akt signaling pathway can inhibit apoptosis. CONCLUSION This review indicates that IL-1β-induced apoptosis plays an important role in pathogenesis and development of pathology of many inflammatory diseases. Elucidating the role of the signaling pathways will aid the development of targeted therapeutic treatments.
Collapse
Affiliation(s)
- Peixuan Wang
- Department of Pediatric Dentistry, Stomatological HospitalSouthern Medical UniversityGuangzhouChina
| | - Hong Qian
- Department of Pediatric Dentistry, Stomatological HospitalSouthern Medical UniversityGuangzhouChina
| | - Manxue Xiao
- Department of Pediatric Dentistry, Stomatological HospitalSouthern Medical UniversityGuangzhouChina
| | - Jingwen Lv
- Department of Pediatric Dentistry, Stomatological HospitalSouthern Medical UniversityGuangzhouChina
| |
Collapse
|
7
|
Song Q, Gao Q, Chen T, Wen T, Wu P, Luo X, Chen QY. FAM3A Ameliorates Brain Impairment Induced by Hypoxia-Ischemia in Neonatal Rat. Cell Mol Neurobiol 2023; 43:251-264. [PMID: 34853925 PMCID: PMC9813043 DOI: 10.1007/s10571-021-01172-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 11/14/2021] [Indexed: 01/12/2023]
Abstract
Hypoxia-ischemia (HI) during crucial periods of brain formation can lead to changes in brain morphology, propagation of neuronal stimuli, and permanent neurodevelopmental impairment, which can have profound effects on cognitive function later in life. FAM3A, a subgroup of family with sequence similarity 3 (FAM3) gene family, is ubiquitously expressed in almost all cells. Overexpression of FAM3A has been evidenced to reduce hyperglycemia via the PI3K/Akt signaling pathway and protect mitochondrial function in neuronal HT22 cells. This study aims to evaluate the protective role of FAM3A in HI-induced brain impairment. Experimentally, maternal rats underwent uterine artery bilateral ligation to induce neonatal HI on day 14 of gestation. At 6 weeks of age, cognitive development assessments including NSS, wire grip, and water maze were carried out. The animals were then sacrificed to assess cerebral mitochondrial function as well as levels of FAM3A, TNF-α and IFN-γ. Results suggest that HI significantly reduced FAM3A expression in rat brain tissues, and that overexpression of FAM3A through lentiviral transduction effectively improved cognitive and motor functions in HI rats as reflected by improved NSS evaluation, cerebral water content, limb strength, as well as spatial learning and memory. At the molecular level, overexpression of FAM3A was able to promote ATP production, balance mitochondrial membrane potential, and reduce levels of pro-inflammatory cytokines TNF-α and IFN-γ. We conclude that FAM3A overexpression may have a protective effect on neuron morphology, cerebral mitochondrial as well as cognitive function. Created with Biorender.com.
Collapse
Affiliation(s)
- Qing Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| | - Qingying Gao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
- The Third Affiliated Hospital of Xi'an Medical University, Xi'an, 710049, Shaanxi, China
| | - Taotao Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Ting Wen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Peng Wu
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China.
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China.
| | - Qiao Yi Chen
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China.
| |
Collapse
|
8
|
Yang L, Xing W, Xiao WZ, Tang L, Wang L, Liu MJ, Dai B. 2,3,5,4'-Tetrahydroxy-stilbene-2- O-beta-d-glucoside induces autophagy-mediated apoptosis in hepatocytes by upregulating miR-122 and inhibiting the PI3K/Akt/mTOR pathway: implications for its hepatotoxicity. PHARMACEUTICAL BIOLOGY 2020; 58:806-814. [PMID: 32881597 PMCID: PMC8641687 DOI: 10.1080/13880209.2020.1803367] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 07/22/2020] [Accepted: 07/26/2020] [Indexed: 06/11/2023]
Abstract
CONTEXT The potential hepatotoxicity of Polygoni Multiflori Radix (PMR) has attracted much attention, but the specific mechanism of inducing hepatotoxicity is still unclear due to the complexity of its components. OBJECTIVE This study investigated the specific mechanism by which 2,3,5,4'-tetrahydroxy-stilbene-2-O-β-d-glucoside (TSG) regulates hepatotoxicity. MATERIALS AND METHODS The toxic effects of TSG (10, 100, 1000 μg/mL) on WRL-68 cells were examined using MTT, flow cytometry, and LDH assay after 24 h of incubation. Untreated cells served as the control. Gene and protein expression levels were determined by quantitative real-time PCR and Western blot, respectively. Immunofluorescence analysis was conducted to investigate the expression of light chain 3 (LC3). Luciferase activity assay was used to assess the targeted regulation of RUNX1 by miR-122. RESULTS The half maximal inhibitory concentration (IC50) of TSG in WRL-68 cells was calculated as 1198.62 μg/mL. TSG (1000 μg/mL) inhibited cell viability and LDH activity and promoted WRL-68 cell apoptosis by inducing autophagy. Subsequent findings showed that TSG induced autophagy and promoted apoptosis in WRL-68 cells by downregulating the levels of p-PI3K, p-Akt, and p-mTOR proteins, while RUNX1 overexpression rescued this inhibition. Additionally, the effect of TSG on hepatocyte apoptosis was reversed by miR-122 knockdown. Furthermore, bioinformatics and dual luciferase reporter assay results indicated that miR-122 targeted RUNX1. DISCUSSION AND CONCLUSIONS Our data demonstrate for the first time that TSG regulates hepatotoxicity, possibly by upregulating miR-122 and inhibiting the RUNX1-mediated PI3K/Akt/mTOR pathway to promote autophagy and induce hepatocyte apoptosis. Further in vivo research is necessary to verify our conclusion.
Collapse
Affiliation(s)
- Lei Yang
- Department of Preparations, The First Hospital of Hunan University of Chinese Medicine, Changsha, P.R. China
| | - Wei Xing
- Department of Intensive Care Medicine, The Third Xiangya Hospital of Central South University, Changsha, P.R. China
| | - Wang-Zhong Xiao
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, P.R. China
| | - Lin Tang
- Department of Preparations, The First Hospital of Hunan University of Chinese Medicine, Changsha, P.R. China
| | - Lu Wang
- Department of Preparations, The First Hospital of Hunan University of Chinese Medicine, Changsha, P.R. China
| | - Meng-Jiao Liu
- Hunan University of Chinese Medicine, Changsha, P.R. China
| | - Bing Dai
- Department of Pharmacy, The First Hospital of Hunan University of Chinese Medicine, Changsha, P.R. China
| |
Collapse
|
9
|
Chen H, Guo M, Yue D, Zhao J, Zhou Y, Chen C, Liang G, Xu L. MicroRNA-7 negatively regulates Toll-like receptor 4 signaling pathway through FAM177A. Immunology 2020; 162:44-57. [PMID: 32852789 DOI: 10.1111/imm.13252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/08/2020] [Accepted: 08/12/2020] [Indexed: 02/06/2023] Open
Abstract
Toll-like receptor (TLR) 4 signalling is critical for innate immunoinflammatory response and widely triggers the development of various types of clinical diseases. MicroRNA-7 (miR-7) is well documented to play an important regulatory role in various biological events. However, the exact role of miR-7 in TLR4 signalling pathway remains to be fully elucidated. In the present study, we found that miR-7 expression in TLR4 signalling-activated bone marrow-derived macrophages (BMDMs) stimulated by LPS was dramatically increased. Importantly, miR-7 deficiency significantly enhanced the production of related inflammatory cytokines including IL-1β, IL-6 and IL-12, as well as TNF-α, on LPS-activated BMDMs, accompanied by elevated transduction of TLR4 signalling including Myd88-dependent and Myd88-independent pathways, whereas miR-7 overexpression significantly decreased the transduction of TLR4 signalling and the production of related inflammatory cytokines. Mechanistically, we identified family with sequence similarity 177, member A (FAM177A) as a novel target molecule of miR-7. Furthermore, down-regulation of FAM177A using RNAi could impair the transduction of TLR4 signalling. Finally, down-regulation of FAM177A also reversed the effect of miR-7 deficiency on TLR4 signalling transduction and production of related inflammatory cytokines on BMDMs. Therefore, we provide the new evidence that miR-7 acts as a novel negative fine-tuner in regulating TLR4 signalling pathways by targeting FAM177A, which might throw light on the basal understanding on the regulatory mechanism of TLR4 signalling and benefit the development of therapeutic strategies against related clinical diseases.
Collapse
Affiliation(s)
- Huizi Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, China.,Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Mengmeng Guo
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, China.,Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Dongxu Yue
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, China.,Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Juanjuan Zhao
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, China.,Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Ya Zhou
- Department of Medical Physics, Zunyi Medical University, Zunyi, China
| | - Chao Chen
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, China.,Department of Immunology, Zunyi Medical University, Zunyi, China
| | - Guiyou Liang
- Department of Cardiovascular Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Department of Cardiovascular Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Lin Xu
- Special Key Laboratory of Gene Detection & Therapy of Guizhou Province, Zunyi, China.,Department of Immunology, Zunyi Medical University, Zunyi, China
| |
Collapse
|
10
|
Fan Z, Liu Y, Shi Z, Deng K, Zhang H, Li Q, Cao S, Li S, Zhang H. MiR-155 promotes interleukin-1β-induced chondrocyte apoptosis and catabolic activity by targeting PIK3R1-mediated PI3K/Akt pathway. J Cell Mol Med 2020; 24:8441-8451. [PMID: 32562373 PMCID: PMC7412691 DOI: 10.1111/jcmm.15388] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 04/01/2020] [Accepted: 04/27/2020] [Indexed: 12/23/2022] Open
Abstract
Osteoarthritis (OA) is a common joint disease characterized by progressive cartilage degradation, in which elevated chondrocyte apoptosis and catabolic activity play an important role. MicroRNA-155 (miR-155) has recently been shown to regulate apoptosis and catabolic activity in some pathological circumstances, yet, whether and how miR-155 is associated with OA pathology remain unexplored. We report here that miR-155 level is significantly up-regulated in human OA cartilage biopsies and also in primary chondrocytes stimulated by interleukin-1β (IL-1β), a pivotal pro-catabolic factor promoting cartilage degradation. Moreover, miR-155 inhibition attenuates and its overexpression promotes IL-1β-induced apoptosis and catabolic activity in chondrocytes in vitro. We also demonstrate that the PIK3R1 (p85α regulatory subunit of phosphoinositide 3-kinase (PI3K)) is a target of miR-155 in chondrocytes, and more importantly, PIK3R1 restoration abrogates miR-155 effects on chondrocyte apoptosis and catabolic activity. Mechanistically, PIK3R1 positively regulates the transduction of PI3K/Akt pathway, and a specific Akt inhibitor reverses miR-155 effects on promoting chondrocyte apoptosis and catabolic activity, phenocopying the results obtained via PIK3R1 knockdown, hence establishing that miR-155 promotes chondrocyte apoptosis and catabolic activity through targeting PIK3R1-mediated PI3K/Akt pathway activation. Altogether, our study discovers novel roles and mechanisms of miR-155 in regulating chondrocyte apoptosis and catabolic activity, providing an implication for therapeutically intervening cartilage degradation and OA progression.
Collapse
Affiliation(s)
- Zhiyong Fan
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Yinghui Liu
- Department of Infectious DiseaseThe Third Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Zhengliang Shi
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Kai Deng
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Hua Zhang
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Qiutong Li
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Shuxing Cao
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Shentai Li
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| | - Hongliang Zhang
- Department of Orthopaedic SurgeryThe Second Hospital of Hebei Medical UniversityShijiazhuangChina
| |
Collapse
|
11
|
Shi X, Han L, Sun T, Zhang F, Ji S, Zhang M, Wang X, Yang W. Silencing UHRF1 enhances cell autophagy to prevent articular chondrocytes from apoptosis in osteoarthritis through PI3K/AKT/mTOR signaling pathway. Biochem Biophys Res Commun 2020; 529:1018-1024. [PMID: 32819559 DOI: 10.1016/j.bbrc.2020.06.032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is a common chronic degenerative joint disease, and chondrocyte apoptosis is one of most important pathological changes of OA pathogenesis. Growing studies have shown that Ubiquitin-like with PHD and RING finger domains 1 (UHRF1) is an important epigenetic regulatory factor that regulates cell proliferation and apoptosis of various tumors, but its role in OA remains ill-defined. In the present study, we found that UHRF1 expression was increased in human OA cartilage tissues, compared with normal cartilage tissues. Interleukin-1β (IL-1β), a major inflammatory cytokine that promotes cartilage degradation in OA, was used to stimulate primary human chondrocytes in vitro. The expression of UHRF1 was also enhanced in IL-1β-induced chondrocytes. Moreover, down-regulation of UHRF1 induced an increase on cell proliferation and autophagy, and a decrease on apoptosis of chondrocytes after IL-1β treatment. Further data indicated that silencing UHRF1 attenuated the up-regulation of IL-1β on phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) signaling pathway in chondrocytes. Then, an activator of PI3K weakened the effect of UHRF1 silencing on cell proliferation, autophagy, apoptosis of IL-1β-induced chondrocytes, and the cell autophagy special inhibitor 3-methyladenine (3-MA) also showed a same impact on UHRF1, hence suggesting that knockdown of UHRF1 enhances cell autophagy to protect chondrocytes from apoptosis in OA through PI3K/AKT/mTOR signaling pathway. In conclusion, our study suggests that UHRF1 may be a potential regulator of chondrocyte apoptosis in the pathogenesis of OA.
Collapse
Affiliation(s)
- Xiaojuan Shi
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lei Han
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Tianshu Sun
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Feng Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Shiying Ji
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Min Zhang
- Department of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaoqing Wang
- Outpatient Department, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Weihong Yang
- Outpatient Department,No.986 Hospital,The Forth Military Medical University, Xi'an, 710054, China.
| |
Collapse
|
12
|
Wen X, Li H, Sun H, Zeng A, Lin R, Zhao J, Zhang Z. MiR-455-3p reduces apoptosis and alleviates degeneration of chondrocyte through regulating PI3K/AKT pathway. Life Sci 2020; 253:117718. [PMID: 32343998 DOI: 10.1016/j.lfs.2020.117718] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 04/13/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Abstract
AIMS This study aimed to explore the functions of miR-455-3p, PTEN, and PI3K/AKT pathway in osteoarthritis. MATERIALS AND METHODS We used the human bone marrow stem cell (BMSC), healthy chondrocytes, osteoarthritis chondrocytes (OA), and the IL-1β/TNF-α-treated chondrocyte model to explore the relationship between miR-455-3p and PTEN. Mimic or inhibitor was used to transfect chondrocytes to determine whether miR-455-3p can regulate PTEN and influence COL2A1 and MMP13. Apoptosis was detected by flow cytometry. A luciferase report was applied to verify the targeted binding. KO mice were applied to investigate PTEN and pAKT expression and the effect on chondrocytes in vivo. KEY FINDINGS MiR-455-3p and PTEN were reverse in chondrogenesis and healthy cartilage versus OA cartilage. Similar trends were noted in IL-1β model. PTEN and MMP13 decreased and COL2A1 increased after overexpressing miR-455-3p, whereas the inhibition showed opposite results. Flow cytometry showed that miR-455-3p could reduce the apoptosis of chondrocytes. The results of luciferase revealed that miR-455-3p could affect fluorescence activity of PTEN by targeting its 3'-UTR. Finally, we found a marked increased in the expression of PTEN in KO mice relative to WT mice, while pAKT levels decreased. SIGNIFICANCE It can be supported that miR-455-3p can reduce the apoptosis of chondrocytes and alleviate OA through regulating PI3K/AKT pathway, which may be expected to be a target for the treatment of osteoarthritis.
Collapse
Affiliation(s)
- Xingzhao Wen
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Hongyi Li
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Hao Sun
- Department of Joint Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Anyu Zeng
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Ruifu Lin
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Jing Zhao
- Department of Medical Imaging, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.
| | - Zhiqi Zhang
- Department of Joint Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China.
| |
Collapse
|