1
|
Chen F, Jiang R, Yu X. Circular RNA circ_0002984 Facilitates the Proliferation and Migration of Ox-LDL-Induced Vascular Smooth Muscle Cells via the Let-7a-5p/KLF5 Pathway. Cardiovasc Toxicol 2024; 24:1253-1267. [PMID: 39181944 DOI: 10.1007/s12012-024-09911-z] [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: 05/07/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024]
Abstract
Circular RNAs (circRNAs) play an important role in the progression of atherosclerosis (AS). This study aimed to explore the exact role and mechanism of circ_0002984 in oxidized low-density lipoprotein (ox-LDL)-mediated human vascular smooth muscle cells (HVSMCs). The model of smooth muscle cell phenotype switching was constructed by treating HVSMCs with ox-LDL. The levels of circ_0002984, let-7a-5p, and kruppel-like factor 5 (KLF5) were measured by quantitative real-time PCR or western blot assay. Cell proliferation, migration, and apoptosis were detected by Cell Counting Kit-8 (CCK-8), EdU staining, wound healing assay, transwell assay, and flow cytometry. The expression of cleaved-caspase-3 and KLF5 was examined by western blot. The relationship between let-7a-5p and circ_0002984 or KLF5 was verified by dual-luciferase reporter assay or RIP assay. The results showed that circ_0002984 and KLF5 were up-regulated, while let-7a-5p was down-regulated in AS patients and ox-LDL-disposed HVSMCs. Silence of circ_0002984 suppressed proliferation and migration, and promoted apoptosis in ox-LDL-stimulated HVSMCs. Moreover, circ_0002984 sponged let-7a-5p to regulate the proliferation, migration, and apoptosis in ox-LDL-resulted HVSMCs. In addition, KLF5 was a target of let-7a-5p and its overexpression reversed the effect of let-7a-5p on the proliferation, migration, and apoptosis in ox-LDL-treated HVSMCs. Also, circ_0002984 positively regulated KLF5 expression by absorbing let-7a-5p. The promotion effect of circ_0002984 on the proliferation and migration of ox-LDL-treated HVSMCs was reversed by KLF5 silencing. Taken together, depletion of circ_0002984 inhibited the proliferation and migration of ox-LDL-stimulated HVSMCs, which might be achieved by modulating the let-7a-5p/KLF5 axis.
Collapse
MESH Headings
- Lipoproteins, LDL/toxicity
- Humans
- Cell Proliferation/drug effects
- RNA, Circular/metabolism
- RNA, Circular/genetics
- Kruppel-Like Transcription Factors/genetics
- Kruppel-Like Transcription Factors/metabolism
- Cell Movement/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/drug effects
- Signal Transduction
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Apoptosis/drug effects
- Cells, Cultured
- Atherosclerosis/pathology
- Atherosclerosis/metabolism
- Atherosclerosis/genetics
- Gene Expression Regulation
Collapse
Affiliation(s)
- Feng Chen
- Department of Cardiovascular Medicine, Lishui People's Hospital, Lishui, Zhejiang, China
| | - Ruilai Jiang
- Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiufeng Yu
- Department of Emergency Medicine, Lishui People's Hospital, No. 1188 Liyang Street, Yanquan Avenue, Liandu District, Lishui, 323050, Zhejiang, China.
| |
Collapse
|
2
|
Wang L, Zhang X, Huang X, Sha X, Li X, Zheng J, Li S, Wei Z, Wu F. Homoplantaginin alleviates high glucose-induced vascular endothelial senescence by inhibiting mtDNA-cGAS-STING pathway via blunting DRP1-mitochondrial fission-VDAC1 axis. FASEB J 2024; 38:e70127. [PMID: 39436199 DOI: 10.1096/fj.202401299rr] [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: 06/09/2024] [Revised: 10/04/2024] [Accepted: 10/10/2024] [Indexed: 10/23/2024]
Abstract
Vascular endothelial senescence is a major risk factor for diabetic vascular complications. Abnormal mitochondrial fission by dynamically related protein 1 (DRP1) accelerates vascular endothelial cell senescence. Homoplantaginin (Hom) is a flavonoid in Salvia plebeia R. Br. with protecting mitochondrial and repairing vascular properties. However, the relevant mechanism of Hom against diabetic vascular endothelial cell senescence remains unclear. Here, we used db/db mice and high glucose (HG)-treated human umbilical vein endothelial cells (HUVECs) to assess the anti-vascular endothelial cell senescence of Hom. We found that Hom inhibited senescence-associated β-galactosidase activity, decreased the levels of senescence markers, and senescence-associated secretory phenotype factors. Additionally, Hom inhibited the expression of cGAS-STING pathway and downstream inflammatory factors. STING inhibitor H-151 delayed endothelial senescence, whereas STING overexpression attenuated the anti-endothelial senescence effect of Hom. Furthermore, we observed that Hom reduced mitochondrial fragmentation and inhibited abnormal mitochondrial fission using transmission electron microscopy. Importantly, Hom has a stronger effect on mitochondrial fission protein than mitochondrial fusion protein, especially downregulated the expression of DRP1. DRP1 inhibitor Mdivi-1 suppressed cGAS-STING pathway and vascular endothelial senescence, yet DRP1 agonist FCCP attenuated the effect of Hom. Surprisingly, Hom blunted abnormal mitochondrial fission mediated by DRP1 mitochondrial localization, suppressed interaction of DRP1 with VDAC1 and prevented VDAC1 oligomerization, which was necessary for mtDNA escape and subsequent cGAS-STING pathway activation. These results revealed a previously unrecognized mechanism that Hom alleviated vascular endothelial senescence by inhibited mtDNA-cGAS-STING signaling pathway via blunting DRP1-mitochondrial fission-VDAC1 axis.
Collapse
Affiliation(s)
- Lei Wang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xueying Zhang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xi Huang
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xiaotong Sha
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xulu Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Jianmei Zheng
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Shitong Li
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zhifeng Wei
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Feihua Wu
- Department of Pharmacology of Chinese Materia Medica, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
3
|
Liu Y, Niu Z, Wang X, Xiu C, Hu Y, Wang J, Lei Y, Yang J. Yiqihuoxue decoction (GSC) inhibits mitochondrial fission through the AMPK pathway to ameliorate EPCs senescence and optimize vascular aging transplantation regimens. Chin Med 2024; 19:143. [PMID: 39402613 PMCID: PMC11479513 DOI: 10.1186/s13020-024-01008-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND During the aging process, the number and functional activity of endothelial progenitor cells (EPCs) are impaired, leading to the unsatisfactory efficacy of transplantation. Previous studies demonstrated that Yiqihuoxue decoction (Ginseng-Sanqi-Chuanxiong, GSC) exerts anti-vascular aging effects. The purpose of this study is to evaluated the effects of GSC on D-galactose (D-gal)induced senescence and the underlying mechanisms. METHODS The levels of cellular senescence-related markers P16, P21, P53, AMPK and p-AMPK were detected by Western blot analysis (WB). SA-β-gal staining was used to evaluate cell senescence. EPCs function was measured by CCK-8, Transwell cell migration and cell adhesion assay. The morphological changes of mitochondria were detected by confocal microscopy. The protein and mRNA expression of mitochondrial fusion fission Drp1, Mff, Fis1, Mfn1, Mfn2 and Opa1 in mitochondria were detect using WB and RT-qPCR. Mitochondrial membrane potential, mtROS and ATP of EPCs were measured using IF. H&E staining was used to observe the pathological changes and IMT of the aorta. The expressions of AGEs, MMP-2 and VEGF in aorta were measured using Immunohistochemical (IHC). The levels of SOD, MDA, NO and ET-1 in serum were detected by SOD, MDA and NO kits. RESULTS In vitro, GSC ameliorated the senescence of EPCs induced by D-gal and reduced the expression of P16, P21 and P53. The mitochondrial morphology of EPCs was restored, the expression of mitochondrial Drp1, Mff and Fis1 protein was decreased, the levels of mtROS and ATP were decreased, and mitochondrial function was improved. Meanwhile, the expression of AMPK and p-AMPK increased. The improvement effects of GSC on aging and mitochondrial morphology and function were were hindered after adding AMPK inhibitor. In vivo, GSC improved EPCs efficiency, ameliorated aortic structural disorder and decreased IMT in aging mice. The serum SOD level increased and MDA level decreased, indicating the improvement of antioxidant capacity. Increased NO content and ET-1 content suggested improvement of vascular endothelial function. The changes observed in SOD and MMP-2 suggested a reduction in vascular stiffness and the degree of vascular damage. The decreased expression of P21 and P53 indicates the delay of vascular senescence.
Collapse
Affiliation(s)
- Yinan Liu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Graduate School of China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Zenghui Niu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xue Wang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chengkui Xiu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yanhong Hu
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Jiali Wang
- Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, 300120, China
| | - Yan Lei
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Jing Yang
- Beijing Key Laboratory of Traditional Chinese Medicine Basic Research on Prevention and Treatment for Major Diseases, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
4
|
Zheng H, Yang L, Huang H, Lin Y, Chen L. Morroniside improves AngII-induced cardiac fibroblast proliferation, migration, and extracellular matrix deposition by blocking p38/JNK signaling pathway through the downregulation of KLF5. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:6611-6621. [PMID: 38472369 PMCID: PMC11422283 DOI: 10.1007/s00210-024-03039-1] [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: 01/16/2024] [Accepted: 03/05/2024] [Indexed: 03/14/2024]
Abstract
Myocardial fibrosis (MF), which is an inevitable pathological manifestation of many cardiovascular diseases in the terminal stage, often contributes to severe cardiac dysfunction and sudden death. Morroniside (MOR) is the main active component of Cornus officinalis with a variety of biological activities. This study was designed to explore the efficacy of MOR in MF and to investigate its pharmacological mechanism. The viability of MOR-treated human cardiac fibroblast (HCF) cells with or without Angiotensin II (AngII) induction was assessed with Cell Counting Kit-8 (CCK-8). The migration of AngII-induced HCF cells was appraised with a transwell assay. Gelatin zymography analysis was adopted to evaluate the activities of MMP2 and MMP9, while immunofluorescence assay was applied for the estimation of Collagen I and Collagen III. By means of western blot, the expressions of migration-, fibrosis-, and p38/c-Jun N-terminal kinase (JNK) signal pathway-related proteins were resolved. The transfection efficacy of oe-Kruppel-like factor 5 (KLF5) was examined with reverse transcription-quantitative PCR (RT-qPCR) and western blot. In this study, it was found that MOR treatment inhibited AngII-induced hyperproliferation, migration, and fibrosis of HCF cells, accompanied with decreased activities of matrix metalloproteinase 2 (MMP2), matrix metalloproteinase 9 (MMP9), connective tissue growth factor (CTGF), Fibronectin, and α-SMA, which were all reversed by KLF5 overexpression. Collectively, MOR exerted protective effects on MF by blocking p38/JNK signal pathway through the downregulation of KLF5.
Collapse
Affiliation(s)
- Haotian Zheng
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China
| | - Linxin Yang
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China
- Department of Ultraphonic Medicine, Fujian Provincial Hospital, Fuzhou, Fujian, 350001, People's Republic of China
| | - Huashang Huang
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China
| | - Yazhou Lin
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China.
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China.
| | - Lin Chen
- The Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, People's Republic of China.
- Department of Cardiology, Fujian Provincial Hospital, No. 134 East Street, Fuzhou, Fujian, 350001, People's Republic of China.
| |
Collapse
|
5
|
Zeng W, Liang Y, Huang S, Zhang J, Mai C, He B, Shi L, Liu B, Li W, Huang X, Li X. Ciprofloxacin Accelerates Angiotensin-II-Induced Vascular Smooth Muscle Cells Senescence Through Modulating AMPK/ROS pathway in Aortic Aneurysm and Dissection. Cardiovasc Toxicol 2024; 24:889-903. [PMID: 39138741 PMCID: PMC11335803 DOI: 10.1007/s12012-024-09892-z] [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: 10/19/2023] [Accepted: 07/03/2024] [Indexed: 08/15/2024]
Abstract
Aortic aneurysm and dissection (AAD) is a cardiovascular disease that poses a severe threat to life and has high morbidity and mortality rates. Clinical and animal-based studies have irrefutably shown that fluoroquinolones, a commonly prescribed antibiotic for treating infections, significantly increase the risk of AAD. Despite this, the precise mechanism by which fluoroquinolones cause AAD remains unclear. Therefore, this study aims to investigate the molecular mechanism and role of Ciprofloxacin definitively-a type of fluoroquinolone antibiotic-in the progression of AAD. Aortic transcriptome data were collected from GEO datasets to detect the genes and pathways expressed differently between healthy donors and AAD patients. Human primary Vascular Smooth Muscle Cells (VSMCs) were isolated from the aorta. After 72 h of exposure to 110ug/ml Ciprofloxacin or 100 nmol/L AngII, either or combined, the senescent cells were identified through SA-β-gal staining. MitoTracker staining was used to examine the morphology of mitochondria in each group. Cellular Reactive Oxygen Species (ROS) levels were measured using MitoSox and DCFH-DA staining. Western blot assay was performed to detect the protein expression level. We conducted an analysis of transcriptome data from both healthy donors and patients with AAD and found that there were significant changes in cellular senescence-related signaling pathways in the latter group. We then isolated and identified human primary VSMCs from healthy donors (control-VSMCs) and patients' (AAD-VSMCs) aortic tissue, respectively. We found that VSMCs from patients exhibited senescent phenotype as compared to control-VSMCs. The higher levels of p21 and p16 and elevated SA-β-gal activity demonstrated this. We also found that pretreatment with Ciprofloxacin promoted angiotensin-II-induced cellular senescence in control-VSMCs. This was evidenced by increased SA-β-gal activity, decreased cell proliferation, and elevation of p21 and p16 protein levels. Additionally, we found that Angiotensin-II (AngII) induced VSMC senescence by promoting ROS generation. We used DCFH-DA and mitoSOX staining to identify that Ciprofloxacin and AngII pretreatment further elevated ROS levels than the vehicle or alone group. Furthermore, JC-1 staining showed that mitochondrial membrane potential significantly declined in the Ciprofloxacin and AngII combination group compared to others. Compared to the other three groups, pretreatment of Ciprofloxacin plus AngII could further induce mitochondrial fission, demonstrated by mitoTracker staining and western blotting assay. Mechanistically, we found that Ciprofloxacin impaired the balance of mitochondrial fission and fusion dynamics in VSMCs by suppressing the phosphorylation of AMPK signaling. This caused mitochondrial dysfunction and ROS generation, thereby elevating AngII-induced cellular senescence. However, treatment with the AMPK activator partially alleviated those effects. Our data indicate that Ciprofloxacin may accelerate AngII-induced VSMC senescence through modulating AMPK/ROS signaling and, subsequently, hasten the progression of AAD.
Collapse
MESH Headings
- Humans
- Cellular Senescence/drug effects
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/pathology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/enzymology
- Aortic Dissection/chemically induced
- Aortic Dissection/pathology
- Aortic Dissection/enzymology
- Aortic Dissection/metabolism
- Signal Transduction/drug effects
- Reactive Oxygen Species/metabolism
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/pathology
- Myocytes, Smooth Muscle/enzymology
- Myocytes, Smooth Muscle/metabolism
- Angiotensin II/toxicity
- Cells, Cultured
- Ciprofloxacin/pharmacology
- AMP-Activated Protein Kinases/metabolism
- Case-Control Studies
- Aortic Aneurysm/chemically induced
- Aortic Aneurysm/pathology
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/enzymology
- Male
- Middle Aged
- Oxidative Stress/drug effects
Collapse
Affiliation(s)
- Weiyue Zeng
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yaowen Liang
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
- Shantou University Medical College, Shantou, China
| | - Shangjun Huang
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jiarui Zhang
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Cong Mai
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Binbin He
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Linli Shi
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Baojuan Liu
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Weifeng Li
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Xiaoran Huang
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| | - Xin Li
- School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Emergency Medicine, China-Algeria Joint Laboratory On Emergeney Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
6
|
Li C, Liu L, Li S, Liu YS. N 6-Methyladenosine in Vascular Aging and Related Diseases: Clinical Perspectives. Aging Dis 2024; 15:1447-1473. [PMID: 37815911 PMCID: PMC11272212 DOI: 10.14336/ad.2023.0924-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/24/2023] [Indexed: 10/12/2023] Open
Abstract
Aging leads to progressive deterioration of the structure and function of arteries, which eventually contributes to the development of vascular aging-related diseases. N6-methyladenosine (m6A) is the most prevalent modification in eukaryotic RNAs. This reversible m6A RNA modification is dynamically regulated by writers, erasers, and readers, playing a critical role in various physiological and pathological conditions by affecting almost all stages of the RNA life cycle. Recent studies have highlighted the involvement of m6A in vascular aging and related diseases, shedding light on its potential clinical significance. In this paper, we comprehensively discuss the current understanding of m6A in vascular aging and its clinical implications. We discuss the molecular insights into m6A and its association with clinical realities, emphasizing its significance in unraveling the mechanisms underlying vascular aging. Furthermore, we explore the possibility of m6A and its regulators as clinical indicators for early diagnosis and prognosis prediction and investigate the therapeutic potential of m6A-associated anti-aging approaches. We also examine the challenges and future directions in this field and highlight the necessity of integrating m6A knowledge into patient-centered care. Finally, we emphasize the need for multidisciplinary collaboration to advance the field of m6A research and its clinical application.
Collapse
Affiliation(s)
- Chen Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Le Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - Shuang Li
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| | - You-Shuo Liu
- Department of Geriatrics, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Institute of Aging and Age-related Disease Research, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Liu ZL, Li Y, Lin YJ, Shi MM, Fu MX, Li ZQ, Ning DS, Zeng XM, Liu X, Cui QH, Peng YM, Zhou XM, Hu YR, Liu JS, Liu YJ, Wang M, Zhang CX, Kong W, Ou ZJ, Ou JS. Aging aggravates aortic aneurysm and dissection via miR-1204-MYLK signaling axis in mice. Nat Commun 2024; 15:5985. [PMID: 39013850 PMCID: PMC11252124 DOI: 10.1038/s41467-024-50036-2] [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: 04/09/2023] [Accepted: 06/25/2024] [Indexed: 07/18/2024] Open
Abstract
The mechanism by which aging induces aortic aneurysm and dissection (AAD) remains unclear. A total of 430 participants were recruited for the screening of differentially expressed plasma microRNAs (miRNAs). We found that miR-1204 is significantly increased in both the plasma and aorta of elder patients with AAD and is positively correlated with age. Cell senescence induces the expression of miR-1204 through p53 interaction with plasmacytoma variant translocation 1, and miR-1204 induces vascular smooth muscle cell (VSMC) senescence to form a positive feedback loop. Furthermore, miR-1204 aggravates angiotensin II-induced AAD formation, and inhibition of miR-1204 attenuates β-aminopropionitrile monofumarate-induced AAD development in mice. Mechanistically, miR-1204 directly targets myosin light chain kinase (MYLK), leading to the acquisition of a senescence-associated secretory phenotype (SASP) by VSMCs and loss of their contractile phenotype. MYLK overexpression reverses miR-1204-induced VSMC senescence, SASP and contractile phenotypic changes, and the decrease of transforming growth factor-β signaling pathway. Our findings suggest that aging aggravates AAD via the miR-1204-MYLK signaling axis.
Collapse
Affiliation(s)
- Ze-Long Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Yan Li
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Yi-Jun Lin
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Mao-Mao Shi
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Meng-Xia Fu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Zhi-Qing Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P.R. China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P.R. China
| | - Da-Sheng Ning
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Xiang-Ming Zeng
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Xiang Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Qing-Hua Cui
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University, Beijing, P.R. China
| | - Yue-Ming Peng
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Xin-Min Zhou
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, P.R. China
| | - Ye-Rong Hu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital of Central South University, Changsha, P.R. China
| | - Jia-Sheng Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Yu-Jia Liu
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
| | - Mian Wang
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China
- Division of Vascular Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Chun-Xiang Zhang
- Department of Pharmacology and Cardiovascular Research Center, Rush Medical College, Rush University Medical Center, Chicago, IL, USA
- Department of Cardiology, Institute of Cardiovascular Research, the Affiliated Hospital, Southwest Medical University, Luzhou, China
| | - Wei Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing, P.R. China.
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, P.R. China.
| | - Zhi-Jun Ou
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China.
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China.
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China.
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China.
- Division of Hypertension and Vascular Diseases, Department of Cardiology, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China.
| | - Jing-Song Ou
- Division of Cardiac Surgery, Cardiovascular Diseases Institute, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China.
- National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China.
- NHC key Laboratory of Assisted Circulation and Vascular Diseases (Sun Yat-sen University), Guangzhou, P.R. China.
- Key Laboratory of Assisted Circulation and Vascular Diseases, Chinese Academy of Medical Sciences, Guangzhou, P.R. China.
- Guangdong Provincial Engineering and Technology Center for Diagnosis and Treatment of Vascular Diseases, Guangzhou, P.R. China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, P.R. China.
| |
Collapse
|
8
|
Zhang H, Zhang K, Gu Y, Tu Y, Ouyang C. Roles and Mechanisms of miRNAs in Abdominal Aortic Aneurysm: Signaling Pathways and Clinical Insights. Curr Atheroscler Rep 2024; 26:273-287. [PMID: 38709435 DOI: 10.1007/s11883-024-01204-8] [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] [Accepted: 04/24/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE OF REVIEW Abdominal aortic aneurysm refers to a serious medical condition that can cause the irreversible expansion of the abdominal aorta, which can lead to ruptures that are associated with up to 80% mortality. Currently, surgical and interventional procedures are the only treatment options available for treating abdominal aortic aneurysm patients. In this review, we focus on the upstream and downstream molecules of the microRNA-related signaling pathways and discuss the roles, mechanisms, and targets of microRNAs in abdominal aortic aneurysm modulation to provide novel insights for precise and targeted drug therapy for the vast number of abdominal aortic aneurysm patients. RECENT FINDINGS Recent studies have highlighted that microRNAs, which are emerging as novel regulators of gene expression, are involved in the biological activities of regulating abdominal aortic aneurysms. Accumulating studies suggested that microRNAs modulate abdominal aortic aneurysm development through various signaling pathways that are yet to be comprehensively summarized. A total of six signaling pathways (NF-κB signaling pathway, PI3K/AKT signaling pathway, MAPK signaling pathway, TGF-β signaling pathway, Wnt signaling pathway, and P53/P21 signaling pathway), and a total of 19 miRNAs are intimately associated with the biological properties of abdominal aortic aneurysm through targeting various essential molecules. MicroRNAs modulate the formation, progression, and rupture of abdominal aortic aneurysm by regulating smooth muscle cell proliferation and phenotype change, vascular inflammation and endothelium function, and extracellular matrix remodeling. Because of the broad crosstalk among signaling pathways, a comprehensive analysis of miRNA-mediated signaling pathways is necessary to construct a well-rounded upstream and downstream regulatory network for future basic and clinical research of AAA therapy.
Collapse
Affiliation(s)
- Haorui Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Ke Zhang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Yuanrui Gu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Yanxia Tu
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, 167 Beilishi Road, Xi Cheng District, Beijing, 100037, China.
| |
Collapse
|
9
|
Lin Z, Bao R, Niu Y, Kong X. KLF5-mediated pyroptosis of airway epithelial cells leads to airway inflammation in asthmatic mice through the miR-182-5p/TLR4 axis. Mol Immunol 2024; 170:9-18. [PMID: 38593669 DOI: 10.1016/j.molimm.2024.03.007] [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: 11/22/2023] [Revised: 03/07/2024] [Accepted: 03/15/2024] [Indexed: 04/11/2024]
Abstract
Asthma is viewed as an airway disease and an inflammatory condition. This study aims to reveal the role of Kruppel-like factor 5 (KLF5)-mediated pyroptosis of airway epithelial cells in airway inflammation in asthma. The asthmatic mouse model was established. The mice were infected with the lentivirus containing sh-KLF5, antagomiR-182-5p, and pc-Toll-like receptor 4 (TLR4). Airway hyperresponsiveness was measured, and the cells in bronchoalveolar lavage fluid (BALF) were sorted and counted. The expression levels of interleukin (IL)-4/IL-13/IL-6/IL-18/IL-1β/NOD-like receptor family pyrin domain containing 3 (NLRP3)/N-gasdermin D (GSDMD-N)/cleaved caspase-1 were detected. The pathological changes in lung tissue were observed. The enrichment of KLF5 in the miR-182-5p promoter region was measured. The binding relationship among KLF5, miR-182-5p, and TLR4 were analyzed. KLF5 was highly expressed in asthmatic mice. Silencing KLF5 improved airway resistance and lung dynamic compliance, reduced the cells in BALF and the expression of IL-4/IL-13/IL-6/NLRP3/GSDMD-N/cleaved caspase-1/IL-18/IL-1β, and alleviated the pathological changes. Mechanistically, KLF5 bonded to the miR-182-5p promoter to inhibit miR-182-5p expression, and miR-182-5p inhibited TLR4. Silencing miR-182-5p or TLR4 overexpression reversed the improvement of silencing KLF5 on airway inflammation and pyroptosis in asthmatic mice. In conclusion, KLF5 inhibited miR-182-5p to promote TLR4 expression, thus aggravating pyroptosis and airway inflammation in asthmatic mice.
Collapse
Affiliation(s)
- Zhi Lin
- Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China.
| | - Rong Bao
- Department of Clinical Laboratory, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Yang Niu
- Department of Respiratory, Shanxi Province Bronchial Asthma Hospital, China
| | - Xiaomei Kong
- Department of Pulmonary and Critical Care Medicine, Shanxi Province Key Laboratory of Respiratory, The First Hospital of Shanxi Medical University, Taiyuan 030001, China.
| |
Collapse
|
10
|
Liu T, Zhang T, Guo C, Liang X, Wang P, Zheng B. Murine double minute 2-mediated estrogen receptor 1 degradation activates macrophage migration inhibitory factor to promote vascular smooth muscle cell dedifferentiation and oxidative stress during thoracic aortic aneurysm progression. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119661. [PMID: 38218386 DOI: 10.1016/j.bbamcr.2024.119661] [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: 09/08/2023] [Revised: 12/19/2023] [Accepted: 01/03/2024] [Indexed: 01/15/2024]
Abstract
Estrogen receptor 1 (ESR1) has been recently demonstrated as a potential diagnostic biomarker for thoracic aortic aneurysm (TAA). However, its precise role in the progression of TAA remains unclear. In this study, TAA models were established in ApoE-knockout mice and primary mouse vascular smooth muscle cells (VSMCs) through treatment with angiotensin (Ang) II. Our findings revealed a downregulation of ESR1 in Ang II-induced TAA mice and VSMCs. Upregulation of ESR1 mitigated expansion and cell apoptosis in the mouse aorta, reduced pathogenetic transformation of VSMCs, and reduced inflammatory infiltration and oxidative stress both in vitro and in vivo. Furthermore, we identified macrophage migration inhibitory factor (MIF) as a biological target of ESR1. ESR1 bound to the MIF promoter to suppress its transcription. Artificial MIF restoration negated the mitigating effects of ESR1 on TAA. Additionally, we discovered that murine double minute 2 (MDM2) was highly expressed in TAA models and mediated protein degradation of ESR1 through ubiquitination modification. Silencing of MDM2 reduced VSMC dedifferentiation and suppressed oxidative stress. However, these effects were reversed upon further silencing of ESR1. In conclusion, this study demonstrates that MDM2 activates MIF by mediating ESR1 degradation, thus promoting VSMC dedifferentiation and oxidative stress during TAA progression.
Collapse
Affiliation(s)
- Tao Liu
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China; Department of Cardiovascular Surgery, Guangxi International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine, Nanning 530001, Guangxi, PR China
| | - Tian Zhang
- Department of Cardiovascular Surgery, Guangxi International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine, Nanning 530001, Guangxi, PR China
| | - Chenfan Guo
- Department of Cardiovascular Surgery, Guangxi International Zhuang Medicine Hospital, Guangxi University of Chinese Medicine, Nanning 530001, Guangxi, PR China
| | - Xiangsen Liang
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, PR China
| | - Pandeng Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China.
| | - Baoshi Zheng
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, PR China.
| |
Collapse
|
11
|
Yuce K, Ozkan AI. The kruppel-like factor (KLF) family, diseases, and physiological events. Gene 2024; 895:148027. [PMID: 38000704 DOI: 10.1016/j.gene.2023.148027] [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: 02/14/2023] [Revised: 11/06/2023] [Accepted: 11/21/2023] [Indexed: 11/26/2023]
Abstract
The Kruppel-Like Factor family of regulatory proteins, which has 18 members, is transcription factors. This family contains zinc finger proteins, regulates the activation and suppression of transcription, and binds to DNA, RNA, and proteins. Klfs related to the immune system are Klf1, Klf2, Klf3, Klf4, Klf6, and Klf14. Klfs related to adipose tissue development and/or glucose metabolism are Klf3, Klf7, Klf9, Klf10, Klf11, Klf14, Klf15, and Klf16. Klfs related to cancer are Klf3, Klf4, Klf5, Klf6, Klf7, Klf8, Klf9, Klf10, Klf11, Klf12, Klf13, Klf14, Klf16, and Klf17. Klfs related to the cardiovascular system are Klf4, Klf5, Klf10, Klf13, Klf14, and Klf15. Klfs related to the nervous system are Klf4, Klf7, Klf8, and Klf9. Klfs are associated with diseases such as carcinogenesis, oxidative stress, diabetes, liver fibrosis, thalassemia, and the metabolic syndrome. The aim of this review is to provide information about the relationship of Klfs with some diseases and physiological events and to guide future studies.
Collapse
Affiliation(s)
- Kemal Yuce
- Selcuk University, Medicine Faculty, Department of Basic Medical Sciences, Physiology, Konya, Turkiye.
| | - Ahmet Ismail Ozkan
- Artvin Coruh University, Medicinal-Aromatic Plants Application and Research Center, Artvin, Turkiye.
| |
Collapse
|
12
|
Li HR, Chen GL, Fang XL, Cai XJ, Xu RL, Li DD, Zhang ZW. Circ_0068481 Affects the Human Pulmonary Artery Smooth Muscle Cells' Progression by miR-361-3p/KLF5 Axis. Am J Hypertens 2024; 37:33-45. [PMID: 37738301 DOI: 10.1093/ajh/hpad028] [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: 09/13/2022] [Revised: 01/04/2023] [Accepted: 06/28/2023] [Indexed: 09/24/2023] Open
Abstract
BACKGROUND Uncontrolled proliferation of pulmonary artery smooth muscle cells (PASMCs) contributes to the pathogenesis of pulmonary arterial hypertension (PAH). In this work, we defined the precise part of circ_0068481 in PASMC proliferation and migration induced by hypoxia. We hypothesized that circ_0068481 enhanced hypoxia-induced PASMC proliferation, invasion, and migration through the microRNA (miR)-361-3p/Krüppel-like factor 5 (KLF5) pathway. METHODS Human PASMCs (hPASMCs) were exposed to hypoxic (3% O2) conditions. Circ_0068481, miR-361-3p, and KLF5 levels were gauged by qRT-PCR and western blot. Cell viability, proliferation, invasion, and migration were detected by XTT, EdU incorporation, transwell, and wound-healing assays, respectively. Dual-luciferase reporter, RNA immunoprecipitation, and RNA pull-down assays were performed to confirm the direct relationship between miR-361-3p and circ_0068481 or KLF5. RESULTS Circ_0068481 expression was increased in the serum of PAH patients and hypoxia-induced hPASMCs. Downregulation of circ_0068481 attenuated hypoxia-induced promotion in hPASMC proliferation, invasion, and migration. Circ_0068481 directly targeted miR-361-3p, and miR-361-3p downregulation reversed the inhibitory effects of circ_0068481 silencing on hypoxia-induced hPASMC proliferation, invasion, and migration. KLF5 was a direct miR-361-3p target, and miR-361-3p upregulation mitigated hypoxia-induced hPASMC proliferation, invasion, and migration by inhibiting KLF5 expression. Moreover, circ_0068481-induced KLF5 expression by binding to miR-361-3p in hypoxic hPASMCs. CONCLUSIONS Circ_0068481 knockdown ameliorated hypoxia-induced hPASMC proliferation, invasion, and migration at least in part through the miR-361-3p/KLF5 axis.
Collapse
Affiliation(s)
- Hai-Rong Li
- Department of Cardiology, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Guan-Liang Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Xiao-Li Fang
- Department of Cardiology, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Xing-Jiu Cai
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Rong-Li Xu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Dong-Dong Li
- Laboratory department, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Zhi-Wei Zhang
- Department of Cardiology, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Department of Pediatric Cardiology, Guangdong Cardiovascular Institute, Guangdong General Hospital, Guangzhou, China
| |
Collapse
|
13
|
Xiong H, Ye J, Luo Q, Li W, Xu N, Yang H. Exosomal EIF5A derived from Lewis lung carcinoma induced adipocyte wasting in cancer cachexia. Cell Signal 2023; 112:110901. [PMID: 37743008 DOI: 10.1016/j.cellsig.2023.110901] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 08/11/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
Cancer cachexia is a systemic inflammation-driven syndrome, characterized by muscle atrophy and adipose tissue wasting, with progressive weight loss leading to serious impairment of physiological function. Extracellular vesicles (EVs) derived from cancer cells play a significant role in adipocyte lipolysis, yet the mechanism remain uneclucidated. In this study, EVs derived from Lewis lung carcinoma (LLC) cells were extracted and characterized. 3T3-L1 and HIB1B adipocytes were cultured with conditioned medium or EVs from LLC, and LLC cells were used to establish a cancer cachexia mouse model. EVs derived from LLC cells were taken up by 3T3-L1 and HIB1B adipocytes, and derived exosomal EIF5A protein-induced lipolysis of adipocytes. High level of EIF5A was expressed in EVs from LLC cells, exosomal EIF5A is linked to lipid metabolism. Elevated expression of EIF5A is associated with shorter overall survival in lung cancer patients. Western blots, glycerol release and Oil red O staining assays were used to evaluate lipolysis of adipocytes. The reduction of lipolysis in 3T3-L1 and HIB1B adipocytes is achieved through silencing EIF5A or treating with pharmacologic inhibitor GC7 in vitro, and suppressing the expression of EIF5A in LLC cells by infected with shRNA or GC7 treatment partly alleviated white and brown adipose tissue lipolysis in vivo. Mechanistically, EIF5A directly binds with G protein-coupled bile acid receptor 1 (GPBAR1) mRNA to promote its translation and then activates cAMP response element binding protein (CREB) signaling pathway to induce lipolysis. This study demonstrates that exosomal EIF5A from LLC cells, with hypusinated EIF5A, has a lipolytic effect on adipocyte and adipose tissues in cancer cachexia model. Exosomal EIF5A could be involved in lipolysis and these findings indicate that a novel regulator and potential target for cachexia treatment.
Collapse
Affiliation(s)
- Hairong Xiong
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiaxin Ye
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qianqian Luo
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wen Li
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ning Xu
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongmei Yang
- Department of Pathogenic Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
14
|
Jadli A, Gomes K, Ballasy N, Wijesuriya T, Belke D, Fedak P, Patel V. Inhibition of smooth muscle cell death by Angiotensin 1-7 protects against abdominal aortic aneurysm. Biosci Rep 2023; 43:BSR20230718. [PMID: 37947205 PMCID: PMC10695742 DOI: 10.1042/bsr20230718] [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: 04/29/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) represents a debilitating vascular disease characterized by aortic dilatation and wall rupture if it remains untreated. We aimed to determine the effects of Ang 1-7 in a murine model of AAA and to investigate the molecular mechanisms involved. Eight- to 10-week-old apolipoprotein E-deficient mice (ApoEKO) were infused with Ang II (1.44 mg/kg/day, s.c.) and treated with Ang 1-7 (0.576 mg/kg/day, i.p.). Echocardiographic and histological analyses showed abdominal aortic dilatation and extracellular matrix remodeling in Ang II-infused mice. Treatment with Ang 1-7 led to suppression of Ang II-induced aortic dilatation in the abdominal aorta. The immunofluorescence imaging exhibited reduced smooth muscle cell (SMC) density in the abdominal aorta. The abdominal aortic SMCs from ApoEKO mice exhibited markedly increased apoptosis in response to Ang II. Ang 1-7 attenuated cell death, as evident by increased SMC density in the aorta and reduced annexin V/propidium iodide-positive cells in flow cytometric analysis. Gene expression analysis for contractile and synthetic phenotypes of abdominal SMCs showed preservation of contractile phenotype by Ang 1-7 treatment. Molecular analyses identified increased mitochondrial fission, elevated cellular and mitochondrial reactive oxygen species (ROS) levels, and apoptosis-associated proteins, including cytochrome c, in Ang II-treated aortic SMCs. Ang 1-7 mitigated Ang II-induced mitochondrial fission, ROS generation, and levels of pro-apoptotic proteins, resulting in decreased cell death of aortic SMCs. These results highlight a critical vasculo-protective role of Ang 1-7 in a degenerative aortic disease; increased Ang 1-7 activity may provide a promising therapeutic strategy against the progression of AAA.
Collapse
Affiliation(s)
- Anshul S. Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Karina P. Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Noura N. Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Tishani Methsala Wijesuriya
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| | - Darrell Belke
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Paul W.M. Fedak
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Vaibhav B. Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
15
|
Lin MJ, Hu SL, Tian Y, Zhang J, Liang N, Sun R, Gong SX, Wang AP. Targeting Vascular Smooth Muscle Cell Senescence: A Novel Strategy for Vascular Diseases. J Cardiovasc Transl Res 2023; 16:1010-1020. [PMID: 36973566 DOI: 10.1007/s12265-023-10377-7] [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: 11/28/2022] [Accepted: 03/13/2023] [Indexed: 03/29/2023]
Abstract
Vascular diseases are a major threat to human health, characterized by high rates of morbidity, mortality, and disability. VSMC senescence contributes to dramatic changes in vascular morphology, structure, and function. A growing number of studies suggest that VSMC senescence is an important pathophysiological mechanism for the development of vascular diseases, including pulmonary hypertension, atherosclerosis, aneurysm, and hypertension. This review summarizes the important role of VSMC senescence and senescence-associated secretory phenotype (SASP) secreted by senescent VSMCs in the pathophysiological process of vascular diseases. Meanwhile, it concludes the progress of antisenescence therapy targeting VSMC senescence or SASP, which provides new strategies for the prevention and treatment of vascular diseases.
Collapse
Affiliation(s)
- Meng-Juan Lin
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Shi-Liang Hu
- Department of Rheumatology, Shaoyang Central Hospital, Shaoyang, 422000, China
| | - Ying Tian
- Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China
| | - Jing Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Na Liang
- Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China
| | - Rong Sun
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China
| | - Shao-Xin Gong
- Department of Pathology, First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Ai-Ping Wang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- Institute of Clinical Research, Department of Clinical Laboratory, Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, Hunan, China.
| |
Collapse
|
16
|
You Y, Chen X, Chen Y, Pang J, Chen Q, Liu Q, Xue H, Zeng Y, Xiao J, Mi J, Tang Y, Ling W. Epigenetic modulation of Drp1-mediated mitochondrial fission by inhibition of S-adenosylhomocysteine hydrolase promotes vascular senescence and atherosclerosis. Redox Biol 2023; 65:102828. [PMID: 37517319 PMCID: PMC10400927 DOI: 10.1016/j.redox.2023.102828] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023] Open
Abstract
AIMS Vascular senescence, which is closely related to epigenetic regulation, is an early pathological condition in cardiovascular diseases including atherosclerosis. Inhibition of S-adenosylhomocysteine hydrolase (SAHH) and the consequent increase of S-adenosylhomocysteine (SAH), a potent inhibitor of DNA methyltransferase, has been associated with an elevated risk of cardiovascular diseases. This study aimed to investigate whether the inhibition of SAHH accelerates vascular senescence and the development of atherosclerosis. METHODS AND RESULTS The case-control study related to vascular aging showed that increased levels of plasma SAH were positively associated with the risk of vascular aging, with an odds ratio (OR) of 3.90 (95% CI, 1.17-13.02). Elevated pulse wave velocity, impaired endothelium-dependent relaxation response, and increased senescence-associated β-galactosidase staining were observed in the artery of SAHH+/- mice at 32 weeks of age. Additionally, elevated expression of p16, p21, and p53, fission morphology of mitochondria, and over-upregulated expression of Drp1 were observed in vascular endothelial cells with SAHH inhibition in vitro and in vivo. Further downregulation of Drp1 using siRNA or its specific inhibitor, mdivi-1, restored the abnormal mitochondrial morphology and rescued the phenotypes of vascular senescence. Furthermore, inhibition of SAHH in APOE-/- mice promoted vascular senescence and atherosclerosis progression, which was attenuated by mdivi-1 treatment. Mechanistically, hypomethylation over the promoter region of DRP1 and downregulation of DNMT1 were demonstrated with SAHH inhibition in HUVECs. CONCLUSIONS SAHH inhibition epigenetically upregulates Drp1 expression through repressing DNA methylation in endothelial cells, leading to vascular senescence and atherosclerosis. These results identify SAHH or SAH as a potential therapeutic target for vascular senescence and cardiovascular diseases.
Collapse
Affiliation(s)
- Yiran You
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Xu Chen
- Department of Molecular, Cellular and Developmental Biology, University of Colorado, Boulder, USA
| | - Yu Chen
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Juan Pang
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Qian Chen
- Department of Cardiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, People's Republic of China; School of Public Health and Management, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Qiannan Liu
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Hongliang Xue
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yupeng Zeng
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jinghe Xiao
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Jiaxin Mi
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Yi Tang
- Department of Nutrition, The First People's Hospital of Zhaoqing, Zhaoqing, China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-Sen University, Guangzhou, People's Republic of China; School of Public Health and Management, Ningxia Medical University, Yinchuan, People's Republic of China; Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, People's Republic of China.
| |
Collapse
|
17
|
Li J, Li X, Song S, Sun Z, Li Y, Yang L, Xie Z, Cai Y, Zhao Y. Mitochondria spatially and temporally modulate VSMC phenotypes via interacting with cytoskeleton in cardiovascular diseases. Redox Biol 2023; 64:102778. [PMID: 37321061 DOI: 10.1016/j.redox.2023.102778] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/06/2023] [Indexed: 06/17/2023] Open
Abstract
Cardiovascular diseases caused by atherosclerosis (AS) seriously endanger human health, which is closely related to vascular smooth muscle cell (VSMC) phenotypes. VSMC phenotypic transformation is marked by the alteration of phenotypic marker expression and cellular behaviour. Intriguingly, the mitochondrial metabolism and dynamics altered during VSMC phenotypic transformation. Firstly, this review combs VSMC mitochondrial metabolism in three aspects: mitochondrial ROS generation, mutated mitochondrial DNA (mtDNA) and calcium metabolism respectively. Secondly, we summarized the role of mitochondrial dynamics in regulating VSMC phenotypes. We further emphasized the association between mitochondria and cytoskelton via presenting cytoskeletal support during mitochondrial dynamics process, and discussed its impact on their respective dynamics. Finally, considering that both mitochondria and cytoskeleton are mechano-sensitive organelles, we demonstrated their direct and indirect interaction under extracellular mechanical stimuli through several mechano-sensitive signaling pathways. We additionally discussed related researches in other cell types in order to inspire deeper thinking and reasonable speculation of potential regulatory mechanism in VSMC phenotypic transformation.
Collapse
Affiliation(s)
- Jingwen Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Xinyue Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Sijie Song
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhengwen Sun
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yuanzhu Li
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Long Yang
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Zhenhong Xie
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yikui Cai
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Yinping Zhao
- Laboratory of Tissue and Cell Biology, Lab Teaching & Management Center, Chongqing Medical University, NO.1 Medical College Road, Yuzhong District, Chongqing, 400016, China.
| |
Collapse
|
18
|
Liao KM, Chen CJ, Luo WJ, Hsu CW, Yu SL, Yang PC, Su KY. Senomorphic effect of diphenyleneiodonium through AMPK/MFF/DRP1 mediated mitochondrial fission. Biomed Pharmacother 2023; 162:114616. [PMID: 37004322 DOI: 10.1016/j.biopha.2023.114616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/22/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
With an aging population and the numerous health impacts associated with old age, the identification of anti-aging drugs has become an important new research direction. Although mitochondria have been recognized to affect aging, anti-aging drugs specifically targeting the mitochondria are less well characterized. In this study, diphenyleneiodonium (DPI) was identified as a potential senomorphic drug that functions by promoting mitochondrial fission. DPI significantly reduced the number of senescence-associated β-galactosidase (SA-β-gal) positive cells and increased the number of proliferating Ki-67 positive cells in BrdU or irradiation stress-induced senescent NIH3T3 cells or IMR90 cells and mouse embryonic fibroblasts (MEFs) replicative senescent cells. Cell cycle arrest genes and senescence-associated secretory phenotype (SASP) factors were downregulated with DPI treatment. In addition, the oxygen consumption rate (OCR) of mitochondrial respiration showed that DPI significantly reduced senescence-associated hyper OCR. Mechanistically, DPI promoted mitochondrial fission by enhancing AMPK/MFF phosphorylation and DRP1 mitochondrial translocation. Inhibition of DRP1 by Mdivi-1 abolished DPI-induced mitochondrial fission and the anti-senescence phenotype. Importantly, Eighty-eight-week-old mice treated with DPI had significantly reduced numbers of SA-β-gal positive cells and reduced expression of cell cycle arrest genes and SASP factors in their livers and kidneys. Pathological and functional assays showed DPI treatment not only reduced liver fibrosis and immune cell infiltration but also improved aged-related physical impairments in aged mice. Taken together, our study identified a potential anti-aging compound that exerts its effects through modulation of mitochondrial morphology.
Collapse
|
19
|
Deng B, Li A, Zhu Y, Zhou Y, Fei J, Miao Y. SHCBP1 contributes to the proliferation and self‑renewal of cervical cancer cells and activation of the NF‑κB signaling pathway through EIF5A. Oncol Lett 2023; 25:246. [PMID: 37153055 PMCID: PMC10161342 DOI: 10.3892/ol.2023.13832] [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: 09/08/2022] [Accepted: 02/24/2023] [Indexed: 05/09/2023] Open
Abstract
Cervical cancer (CC) is the most common human papillomavirus-related disease. Continuous activation of the NF-κB signaling pathway has been observed in CC. SHC binding and spindle associated 1 (SHCBP1) contributes to tumorigenesis and activation of the NF-κB pathway in multiple cancer types, while its function in CC remains unclear. In the present study, three Gene Expression Omnibus datasets were used to identify differentially expressed genes (DEGs) in CC. Loss- and gain-of-function experiments were performed using stable SHCBP1-silenced and SHCBP1-overexpressing CC cells. To further explore the molecular mechanism of SHCBP1 in CC, small interfering RNA targeting eukaryotic translation initiation factor 5A (EIF5A) was transfected into stable SHCBP1-overexpressing CC cells. The results demonstrated that SHCBP1 was an upregulated DEG in CC tissues compared with healthy control cervical tissues. Functional experiments revealed the pro-proliferative and pro-stemness role of SHCBP1 in CC cells (CaSki and SiHa cells), in vitro. Furthermore, the NF-κB signaling pathway in CC cells was activated by SHCBP1. Increases in cell proliferation, stemness and activation of NF-κB, induced by SHCBP1 overexpression in CC cells, were reversed by EIF5A knockdown. Taken together, the results indicated that SHCBP1 serves an important role in regulation of CC cell proliferation, self-renewal and activation of NF-κB via EIF5A. The present study demonstrated a potential molecular mechanism underlying the progression of CC.
Collapse
Affiliation(s)
- Boya Deng
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
- Correspondence to: Dr Boya Deng, Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, 88 Jiefang Road, Shangcheng, Hangzhou, Zhejiang 310009, P.R. China, E-mail:
| | - Ailin Li
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Ying Zhu
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Yingying Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Jing Fei
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang 310009, P.R. China
| | - Yuan Miao
- Department of Pathology, The College of Basic Medicine Science and The First Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
20
|
Wang D, Jia L, Zhao C, Wang H, Dai Z, Jing Y, Jiang B, Xin S. Mitochondrial quality control in abdominal aortic aneurysm: From molecular mechanisms to therapeutic strategies. FASEB J 2023; 37:e22969. [PMID: 37184038 DOI: 10.1096/fj.202202158rr] [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: 12/28/2022] [Revised: 03/20/2023] [Accepted: 05/01/2023] [Indexed: 05/16/2023]
Abstract
Mitochondria are the energy supply sites of cells and are crucial for eukaryotic life. Mitochondrial dysfunction is involved in the pathogenesis of abdominal aortic aneurysm (AAA). Multiple mitochondrial quality control (MQC) mechanisms, including mitochondrial DNA repair, biogenesis, antioxidant defense, dynamics, and autophagy, play vital roles in maintaining mitochondrial homeostasis under physiological and pathological conditions. Abnormalities in these mechanisms may induce mitochondrial damage and dysfunction leading to cell death and tissue remodeling. Recently, many clues suggest that dysregulation of MQC is closely related to the pathogenesis of AAA. Therefore, specific interventions targeting MQC mechanisms to maintain and restore mitochondrial function have become promising therapeutic methods for the prevention and treatment of AAA.
Collapse
Affiliation(s)
- Ding Wang
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Longyuan Jia
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Chengdong Zhao
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Huitao Wang
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Zhengnan Dai
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Yuchen Jing
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Bo Jiang
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| | - Shijie Xin
- Department of Vascular Surgery, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning Province, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of aortic aneurysm, Shenyang, Liaoning Province, China
| |
Collapse
|
21
|
Gonzalez-Menendez P, Phadke I, Olive ME, Joly A, Papoin J, Yan H, Galtier J, Platon J, Kang SWS, McGraw KL, Daumur M, Pouzolles M, Kondo T, Boireau S, Paul F, Young DJ, Lamure S, Mirmira RG, Narla A, Cartron G, Dunbar CE, Boyer-Clavel M, Porat-Shliom N, Dardalhon V, Zimmermann VS, Sitbon M, Dever TE, Mohandas N, Da Costa L, Udeshi ND, Blanc L, Kinet S, Taylor N. Arginine metabolism regulates human erythroid differentiation through hypusination of eIF5A. Blood 2023; 141:2520-2536. [PMID: 36735910 PMCID: PMC10273172 DOI: 10.1182/blood.2022017584] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 01/20/2023] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Metabolic programs contribute to hematopoietic stem and progenitor cell (HSPC) fate, but it is not known whether the metabolic regulation of protein synthesis controls HSPC differentiation. Here, we show that SLC7A1/cationic amino acid transporter 1-dependent arginine uptake and its catabolism to the polyamine spermidine control human erythroid specification of HSPCs via the activation of the eukaryotic translation initiation factor 5A (eIF5A). eIF5A activity is dependent on its hypusination, a posttranslational modification resulting from the conjugation of the aminobutyl moiety of spermidine to lysine. Notably, attenuation of hypusine synthesis in erythroid progenitors, by the inhibition of deoxyhypusine synthase, abrogates erythropoiesis but not myeloid cell differentiation. Proteomic profiling reveals mitochondrial translation to be a critical target of hypusinated eIF5A, and accordingly, progenitors with decreased hypusine activity exhibit diminished oxidative phosphorylation. This affected pathway is critical for eIF5A-regulated erythropoiesis, as interventions augmenting mitochondrial function partially rescue human erythropoiesis under conditions of attenuated hypusination. Levels of mitochondrial ribosomal proteins (RPs) were especially sensitive to the loss of hypusine, and we find that the ineffective erythropoiesis linked to haploinsufficiency of RPS14 in chromosome 5q deletions in myelodysplastic syndrome is associated with a diminished pool of hypusinated eIF5A. Moreover, patients with RPL11-haploinsufficient Diamond-Blackfan anemia as well as CD34+ progenitors with downregulated RPL11 exhibit a markedly decreased hypusination in erythroid progenitors, concomitant with a loss of mitochondrial metabolism. Thus, eIF5A-dependent protein synthesis regulates human erythropoiesis, and our data reveal a novel role for RPs in controlling eIF5A hypusination in HSPCs, synchronizing mitochondrial metabolism with erythroid differentiation.
Collapse
Affiliation(s)
- Pedro Gonzalez-Menendez
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Ira Phadke
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
- Pediatric Oncology Branch, National Cancer Institute (NCI), Center for Cancer Research (CCR), National Institutes of Health (NIH), Bethesda, MD
| | - Meagan E. Olive
- Proteomics Platform, Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Axel Joly
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Julien Papoin
- Feinstein Institute for Medical Research, Manhasset, NY
- EA4666 HEMATIM, Université Picardie Jules Verne, Amiens, France
| | | | - Jérémy Galtier
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Jessica Platon
- EA4666 HEMATIM, Université Picardie Jules Verne, Amiens, France
| | | | - Kathy L. McGraw
- Laboratory of Receptor Biology and Gene Expression, NCI, CCR, NIH, Bethesda, MD
| | - Marie Daumur
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Marie Pouzolles
- Pediatric Oncology Branch, National Cancer Institute (NCI), Center for Cancer Research (CCR), National Institutes of Health (NIH), Bethesda, MD
| | - Taisuke Kondo
- Pediatric Oncology Branch, National Cancer Institute (NCI), Center for Cancer Research (CCR), National Institutes of Health (NIH), Bethesda, MD
| | - Stéphanie Boireau
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Franciane Paul
- Department of Clinical Hematology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - David J. Young
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Sylvain Lamure
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Department of Clinical Hematology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | | | - Anupama Narla
- Division of Pediatric Hematology/Oncology, Stanford University, Stanford, CA
| | - Guillaume Cartron
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Department of Clinical Hematology, Centre Hospitalier Universitaire de Montpellier, Montpellier, France
| | - Cynthia E. Dunbar
- Translational Stem Cell Biology Branch, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD
| | - Myriam Boyer-Clavel
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | | | - Valérie Dardalhon
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Valérie S. Zimmermann
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
| | - Marc Sitbon
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Thomas E. Dever
- Section on Protein Biosynthesis, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD
| | | | - Lydie Da Costa
- Laboratory of Excellence GR-Ex, Paris, France
- EA4666 HEMATIM, Université Picardie Jules Verne, Amiens, France
- Service d'Hématologie Biologique (Hematology Diagnostic Laboratory), Assistance Publique–Hôpitaux de Paris, Robert Debr Hôpital, Paris, France
- Paris Cité University, Paris, France
| | - Namrata D. Udeshi
- Proteomics Platform, Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Lionel Blanc
- Feinstein Institute for Medical Research, Manhasset, NY
| | - Sandrina Kinet
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
| | - Naomi Taylor
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, Centre National de la Recherche Scientifique (CNRS), Montpellier, France
- Laboratory of Excellence GR-Ex, Paris, France
- Pediatric Oncology Branch, National Cancer Institute (NCI), Center for Cancer Research (CCR), National Institutes of Health (NIH), Bethesda, MD
| |
Collapse
|
22
|
Zhang Y, Huang X, Sun T, Shi L, Liu B, Hong Y, Fu QL, Zhang Y, Li X. MicroRNA-19b-3p dysfunction of mesenchymal stem cell-derived exosomes from patients with abdominal aortic aneurysm impairs therapeutic efficacy. J Nanobiotechnology 2023; 21:135. [PMID: 37101174 PMCID: PMC10131394 DOI: 10.1186/s12951-023-01894-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 04/13/2023] [Indexed: 04/28/2023] Open
Abstract
Senescence of vascular smooth muscle cells (VSMCs) contributes to the formation of abdominal aortic aneurysm (AAA). Although mesenchymal stem cell exosomes (MSC-EXO) have been confirmed to restrict the development of AAA, their biological activity depends largely on the physiological state of the MSCs. This study aimed to compare the effects of adipose-derived MSC-EXO from healthy donors (HMEXO) and AAA patients (AMEXO) on senescence of VSMCs in AAA and explore the underlying mechanisms. An ApoE-/- mouse model of AAA was used to investigate the therapeutic effects of HMEXO, AMEXO or miR-19b-3p-AMEXO on AAA development. This in vitro model of AAA was established by treating VSMCs with Ang II (Angiotensin II). The senescence of VSMCs was determined by senescence-associated β-galactosidase (SA-β-gal) staining. The morphology of mitochondria in VSMCs was examined by MitoTracker staining. HMEXO exhibited superior capacity compared with AMEXO to inhibit VSMC senescence and attenuate AAA formation in Ang II-treated ApoE-/- mice. In vitro, both AMEXO and HMEXO inhibited Ang II-induced VSMC senescence via downregulation of mitochondrial fission. Notably, compared with HMEXO, the ability of AMEXO to inhibit VSMC senescence was significantly decreased. miRNA sequencing and the expression of miR-19b-3p was significantly decreased in AMEXO compared with HMEXO. Luciferase assay suggested that MST4 (Mammalian sterile-20-like kinase 4) is a potential target of miR-19b-3p. Mechanistically, miR-19b-3p in HMEXO ameliorated VSMC senescence by inhibiting mitochondrial fission via regulation of the MST4/ERK/Drp1 signaling pathway. Overexpression of miR-19b-3p in AMEXO improved their beneficial effect on AAA formation. Our study reveals that MSC-exosomal miR-19b-3p exerts protective effects against Ang II-induced AAA and VSMC senescence via regulation of the MST4/ERK/Drp1 pathway. The pathological state of AAA patients alters the miRNA components of AMEXO and impairs their therapeutic benefits.
Collapse
Affiliation(s)
- Yuxiao Zhang
- School of Medicine, South China University of Technology, Guangzhou, China
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaoran Huang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Tucheng Sun
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Linli Shi
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Baojuan Liu
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Yimei Hong
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China
| | - Qing-Ling Fu
- Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuelin Zhang
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| | - Xin Li
- School of Medicine, South China University of Technology, Guangzhou, China.
- Department of Emergency Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
23
|
Metabolic landscape in cardiac aging: insights into molecular biology and therapeutic implications. Signal Transduct Target Ther 2023; 8:114. [PMID: 36918543 PMCID: PMC10015017 DOI: 10.1038/s41392-023-01378-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 02/06/2023] [Accepted: 02/20/2023] [Indexed: 03/16/2023] Open
Abstract
Cardiac aging is evident by a reduction in function which subsequently contributes to heart failure. The metabolic microenvironment has been identified as a hallmark of malignancy, but recent studies have shed light on its role in cardiovascular diseases (CVDs). Various metabolic pathways in cardiomyocytes and noncardiomyocytes determine cellular senescence in the aging heart. Metabolic alteration is a common process throughout cardiac degeneration. Importantly, the involvement of cellular senescence in cardiac injuries, including heart failure and myocardial ischemia and infarction, has been reported. However, metabolic complexity among human aging hearts hinders the development of strategies that targets metabolic susceptibility. Advances over the past decade have linked cellular senescence and function with their metabolic reprogramming pathway in cardiac aging, including autophagy, oxidative stress, epigenetic modifications, chronic inflammation, and myocyte systolic phenotype regulation. In addition, metabolic status is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and chronic inflammation. However, further elucidation of the metabolism involvement in cardiac degeneration is still needed. Thus, deciphering the mechanisms underlying how metabolic reprogramming impacts cardiac aging is thought to contribute to the novel interventions to protect or even restore cardiac function in aging hearts. Here, we summarize emerging concepts about metabolic landscapes of cardiac aging, with specific focuses on why metabolic profile alters during cardiac degeneration and how we could utilize the current knowledge to improve the management of cardiac aging.
Collapse
|
24
|
Zhu T, Hu Q, Yuan Y, Yao H, Zhang J, Qi J. Mitochondrial dynamics in vascular remodeling and target-organ damage. Front Cardiovasc Med 2023; 10:1067732. [PMID: 36860274 PMCID: PMC9970102 DOI: 10.3389/fcvm.2023.1067732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Vascular remodeling is the pathological basis for the development of many cardiovascular diseases. The mechanisms underlying endothelial cell dysfunction, smooth muscle cell phenotypic switching, fibroblast activation, and inflammatory macrophage differentiation during vascular remodeling remain elusive. Mitochondria are highly dynamic organelles. Recent studies showed that mitochondrial fusion and fission play crucial roles in vascular remodeling and that the delicate balance of fusion-fission may be more important than individual processes. In addition, vascular remodeling may also lead to target-organ damage by interfering with the blood supply to major body organs such as the heart, brain, and kidney. The protective effect of mitochondrial dynamics modulators on target-organs has been demonstrated in numerous studies, but whether they can be used for the treatment of related cardiovascular diseases needs to be verified in future clinical studies. Herein, we summarize recent advances regarding mitochondrial dynamics in multiple cells involved in vascular remodeling and associated target-organ damage.
Collapse
Affiliation(s)
- Tong Zhu
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingxun Hu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University, School of Medicine, Shanghai University, Shanghai, China,Shanghai Engineering Research Center of Organ Repair, School of Medicine, Shanghai University, Shanghai, China
| | - Yanggang Yuan
- Department of Nephrology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Huijuan Yao
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian Zhang
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,Jian Zhang,
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China,*Correspondence: Jia Qi,
| |
Collapse
|
25
|
Xia Y, Zhang X, An P, Luo J, Luo Y. Mitochondrial Homeostasis in VSMCs as a Central Hub in Vascular Remodeling. Int J Mol Sci 2023; 24:ijms24043483. [PMID: 36834896 PMCID: PMC9961025 DOI: 10.3390/ijms24043483] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Vascular remodeling is a common pathological hallmark of many cardiovascular diseases. Vascular smooth muscle cells (VSMCs) are the predominant cell type lining the tunica media and play a crucial role in maintaining aortic morphology, integrity, contraction and elasticity. Their abnormal proliferation, migration, apoptosis and other activities are tightly associated with a spectrum of structural and functional alterations in blood vessels. Emerging evidence suggests that mitochondria, the energy center of VSMCs, participate in vascular remodeling through multiple mechanisms. For example, peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α)-mediated mitochondrial biogenesis prevents VSMCs from proliferation and senescence. The imbalance between mitochondrial fusion and fission controls the abnormal proliferation, migration and phenotypic transformation of VSMCs. Guanosine triphosphate-hydrolyzing enzymes, including mitofusin 1 (MFN1), mitofusin 2 (MFN2), optic atrophy protein 1 (OPA1) and dynamin-related protein 1 (DRP1), are crucial for mitochondrial fusion and fission. In addition, abnormal mitophagy accelerates the senescence and apoptosis of VSMCs. PINK/Parkin and NIX/BINP3 pathways alleviate vascular remodeling by awakening mitophagy in VSMCs. Mitochondrial DNA (mtDNA) damage destroys the respiratory chain of VSMCs, resulting in excessive ROS production and decreased ATP levels, which are related to the proliferation, migration and apoptosis of VSMCs. Thus, maintaining mitochondrial homeostasis in VSMCs is a possible way to relieve pathologic vascular remodeling. This review aims to provide an overview of the role of mitochondria homeostasis in VSMCs during vascular remodeling and potential mitochondria-targeted therapies.
Collapse
|
26
|
Liu Y, Peng L, Chen J, Chen L, Wu Y, Cheng M, Chen M, Ye X, Jin Y. EIF5A2 specifically regulates the transcription of aging-related genes in human neuroblastoma cells. BMC Geriatr 2023; 23:83. [PMID: 36750933 PMCID: PMC9906866 DOI: 10.1186/s12877-023-03793-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Post-transcriptional regulation plays a critical role in controlling biological processes such as aging. Previous studies have shown that eukaryotic initiation factor 5A (EIF5A) might play a crucial role in aging. It is unknown whether EIF5A2, a second isoform of EIF5A, could impact aging through post-transcriptional regulation. METHODS In the present study, EIF5A2 overexpression (EIF5A2-OE) was induced in SH-SY5Y cells. RNA-seq, bioinformatics analysis and RT-qPCR validation experiments were then performed to explore the molecular mechanism of EIF5A2-mediated transcriptional regulation. Cell viability, proportion of senescent cells and the cell cycle were respectively determined by Cell Counting Kit-8, SA-β‑galactosidase and flow cytometry to evaluate the cell senescence. RESULTS A total of 190 downregulated and 126 upregulated genes related to EIF5A2-OE were identified. Genes closely related to cellular aging processes such as unfolded protein response (UPR), cell adhesion and calcium signaling pathway were under global transcriptional regulation. Moreover, EIF5A2-OE promoted the viability of SH-SY5Y cells and reduced cell senescence in vitro. Among 30 genes with the most significant expression differences in EIF5A2-OE cells, we identified eight genes, including ASNS, ATF3, ATF4, CEBPB, DDIT3, HERPUD1, HSPA5 and XBP1, enriched in the UPR. Through EIF5A2-tanscription factors (TFs)-targets regulation network in EIF5A2-OE cells, we found three TFs, BHLHE40, RHOXF1 and TBX20, that targeted at these eight UPR-related genes. Verification test via the published database of human glial cell tissue showed only BHLHE40 and RHOXF1 were significantly associated with EIF5A2. CONCLUSIONS Our findings suggest that EIF5A2 may alleviate cell senescence in vitro and mediate UPR-related genes via specific TFs. Thus, EIF5A2 could function as a regulator of aging via the regulation of transcription, which greatly expands the current understanding of the mechanisms of EIF5A2-mediated gene regulation.
Collapse
Affiliation(s)
- Yuwei Liu
- grid.49470.3e0000 0001 2331 6153Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China ,grid.49470.3e0000 0001 2331 6153Department of General Practice, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China
| | - Li Peng
- grid.49470.3e0000 0001 2331 6153Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China ,grid.49470.3e0000 0001 2331 6153Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China
| | - Jing Chen
- grid.49470.3e0000 0001 2331 6153Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China
| | - Ling Chen
- grid.49470.3e0000 0001 2331 6153Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China
| | - Ying Wu
- grid.49470.3e0000 0001 2331 6153Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China
| | - Mengxin Cheng
- grid.49470.3e0000 0001 2331 6153Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China
| | - Min Chen
- grid.49470.3e0000 0001 2331 6153Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei China
| | - Xujun Ye
- Department of Internal Medicine and Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
| | - Yalei Jin
- Department of General Practice, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
27
|
Guo J, Niu K, Ma BF, Sun LN, Fang QW, An JX. Electroacupuncture ameliorates surgery-induced spatial memory deficits by promoting mitophagy in rats. ANNALS OF TRANSLATIONAL MEDICINE 2023; 11:74. [PMID: 36819507 PMCID: PMC9929787 DOI: 10.21037/atm-22-6262] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/10/2023] [Indexed: 01/30/2023]
Abstract
Background This study sought to explore the mechanism underlying the therapeutic effects of electroacupuncture (EA) on spatial memory deficits caused by surgery. Methods Hepatic apex resection was performed under propofol-based total intravenous anesthesia. Male Sprague-Dawley rats were subjected to EA treatment or EA + mitochondrial division inhibitor-1 (mdivi-1) treatment once a day for three consecutive days after surgery. The Morris water maze test was used to evaluate the spatial memory of the rats after surgery. Tissue from the hippocampus of each rat was frozen and used for transcriptomic and proteomic analyses to identify potential targets for EA treatment. Western blotting was used to confirm the protein expression levels. The levels of reactive oxygen species (ROS) and adenosine triphosphate (ATP) were detected using commercial kits. The rat mitochondria were then isolated, and the activity of mitochondrial complex V was assessed. Results EA attenuated surgery-induced spatial memory deficits on postoperative day 3, while these effects were reversed by treatment with the mdivi-1 (P<0.05). Ribonucleic acid (RNA)-sequencing revealed that EA upregulated multiple metabolic pathways and the phosphatidylinositol 3‑kinas/protein kinase B signaling pathway. The proteomic and western blotting results suggested that the EA treatment substantially downregulated coiled-coil-helix-coiled-coil-helix domain containing 3 (ChChd3) expression in the hippocampus. The EA treatment significantly increased the autophagy-related protein levels, including phosphatase and tensin homolog-induced kinase 1, Parkin, MAP1LC3 (LC3), and Beclin1, and inhibited the production of ROS and inflammatory cytokine interleukin-1β in the hippocampus (P<0.05). Conclusions These results suggest that EA ameliorates postoperative spatial memory deficits and protects hippocampus from oxidative stress and inflammation through enhanced autophagy in an animal model of perioperative neurocognitive disorders (PNDs).
Collapse
Affiliation(s)
- Jian Guo
- School of Anesthesiology, Weifang Medical University, Weifang, China;,Department of Anesthesiology, Pain and Sleep Medicine, Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China;,Department of Anesthesiology, Yan’an People’s Hospital, Yan’an, China
| | - Kun Niu
- Department of Anesthesiology, Pain and Sleep Medicine, Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China;,Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Anesthesiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bao-Feng Ma
- School of Anesthesiology, Weifang Medical University, Weifang, China;,Department of Anesthesiology, Pain and Sleep Medicine, Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Li-Na Sun
- School of Anesthesiology, Weifang Medical University, Weifang, China;,Department of Anesthesiology, Pain and Sleep Medicine, Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Qi-Wu Fang
- Department of Anesthesiology, Pain and Sleep Medicine, Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China
| | - Jian-Xiong An
- School of Anesthesiology, Weifang Medical University, Weifang, China;,Department of Anesthesiology, Pain and Sleep Medicine, Aviation General Hospital of China Medical University and Beijing Institute of Translational Medicine, Chinese Academy of Sciences, Beijing, China;,Department of Anesthesiology, Pain and Sleep Medicine, The Affiliated Hospital of Weifang Medical University, Weifang, China
| |
Collapse
|
28
|
Li YJ, Jin X, Li D, Lu J, Zhang XN, Yang SJ, Zhao YX, Wu M. New insights into vascular aging: Emerging role of mitochondria function. Biomed Pharmacother 2022; 156:113954. [DOI: 10.1016/j.biopha.2022.113954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/29/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022] Open
|
29
|
Jiang X, Loayza-Puch F. Roles of eIF5A in the immunosurveillance of cellular senescence. Cancer Biol Med 2022; 19:j.issn.2095-3941.2022.0408. [PMID: 36259416 PMCID: PMC9724223 DOI: 10.20892/j.issn.2095-3941.2022.0408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Affiliation(s)
- Xiangli Jiang
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, Tianjin’s Clinical Research Center for Cancer, Tianjin 300060, China,Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Fabricio Loayza-Puch
- Translational Control and Metabolism, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany,Correspondence to: Fabricio Loayza-Puch, E-mail:
| |
Collapse
|
30
|
Li Y, Guo S, Zhao Y, Li R, Li Y, Qiu C, Xiao L, Gong K. EZH2 Regulates ANXA6 Expression via H3K27me3 and Is Involved in Angiotensin II-Induced Vascular Smooth Muscle Cell Senescence. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4838760. [PMID: 36160712 PMCID: PMC9492406 DOI: 10.1155/2022/4838760] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/14/2022] [Accepted: 08/01/2022] [Indexed: 02/05/2023]
Abstract
Objectives Abdominal aortic aneurysm (AAA) has a high risk of rupture of the aorta and is one of the leading causes of death in older adults. This study is aimed at confirming the influence and mechanism of the abnormally expressed ANXA6 gene in AAA. Methods Clinical samples were collected for proteome sequencing to screen for differentially expressed proteins. An Ang II-induced vascular smooth muscle cell (VSMC) aging model as well as an AAA animal model was used. Using RT-qPCR to detect the mRNA levels of EZH2, ANXA6, IK-6, and IL-8 in cells and tissues were assessed. Western blotting and immunohistochemistry staining were used apply for the expression of associated proteins in cells and tissues. SA-β-gal staining, flow cytometry, and DHE staining were used to detect senescent cells and the level of ROS. The cell cycle was assessed by flow cytometry. Arterial pathology was observed by HE staining. The aging of VSMCs in arterial tissue was assessed by coimmunofluorescence for α-SMA and p53. Results There were 24 differentially expressed proteins in the AAA clinical samples, including 10 upregulated protein and 14 downregulated protein, and the differential expression of ANXA6 was associated with vascular disease. Our study found that ANXA6 was highly expressed and EZH2 was lowly expressed in an Ang II-induced VSMC aging model. Knockdown of ANXA6 or overexpression of EZH2 inhibited Ang II-induced ROS, inhibited cell senescence, decreased Ang II evoked G1 arrest, and increased cells in G2 phase, while overexpression of ANXA6 played the opposite role. Overexpression of EZH2 inhibited ANXA6 expression by increasing H3K27me3 modification at the ANXA6 promoter. Simultaneous overexpression of EZH2 and the protective effect of EZH2 on cell senescence were partially reversed by ANXA6. Similarly, ANXA6 was highly expressed and EZH2 was lowly expressed in an Ang II-induced AAA animal model. Knockdown of ANXA6 and overexpression of EZH2 alleviated Ang II-induced VSMC senescence and inhibited AAA progression, while simultaneous overexpression of EZH2 and ANXA6 partially reversed the protective effect of EZH2 on AAA. Conclusion EZH2 regulates the ANXA6 promoter H3K27me3 modification, inhibits ANXA6 expression, alleviates Ang II-induced VSMC senescence, and inhibits AAA progression.
Collapse
Affiliation(s)
- Yuejin Li
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Shikui Guo
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Yingpeng Zhao
- Department of Hepatic-Biliary-Pancreatic Surgery, The First Hospital of Kunming (The Calmette Hospital), Kunming, Yunnan 650224, China
| | - Rougang Li
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Yu Li
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Changtao Qiu
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Le Xiao
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| | - Kunmei Gong
- Department of General Surgery, The First People's Hospital of Yunnan Province (The Affiliated Hospital of Kunming University of Science and Technology), Kunming, Yunnan 650032, China
| |
Collapse
|
31
|
Wang D, Hao X, Jia L, Jing Y, Jiang B, Xin S. Cellular senescence and abdominal aortic aneurysm: From pathogenesis to therapeutics. Front Cardiovasc Med 2022; 9:999465. [PMID: 36187019 PMCID: PMC9515360 DOI: 10.3389/fcvm.2022.999465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 08/15/2022] [Indexed: 01/10/2023] Open
Abstract
As China’s population enters the aging stage, the threat of abdominal aortic aneurysm (AAA) mainly in elderly patients is becoming more and more serious. It is of great clinical significance to study the pathogenesis of AAA and explore potential therapeutic targets. The purpose of this paper is to analyze the pathogenesis of AAA from the perspective of cellular senescence: on the basis of clear evidence of cellular senescence in aneurysm wall, we actively elucidate specific molecular and regulatory pathways, and to explore the targeted drugs related to senescence and senescent cells eliminate measures, eventually improve the health of patients with AAA and prolong the life of human beings.
Collapse
Affiliation(s)
- Ding Wang
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, Liaoning, China
| | - Xinyu Hao
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, Liaoning, China
| | - Longyuan Jia
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, Liaoning, China
| | - Yuchen Jing
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, Liaoning, China
| | - Bo Jiang
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, Liaoning, China
| | - Shijie Xin
- Department of Vascular Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
- Key Laboratory of Pathogenesis, Prevention and Therapeutics of Aortic Aneurysm, Shenyang, Liaoning, China
- *Correspondence: Shijie Xin,
| |
Collapse
|
32
|
A novel mutation located in the intermembrane space domain of AFG3L2 causes dominant optic atrophy through decreasing the stability of the encoded protein. Cell Death Dis 2022; 8:361. [PMID: 35970831 PMCID: PMC9378676 DOI: 10.1038/s41420-022-01160-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 11/18/2022]
Abstract
Dominant optic atrophy (DOA) is the most common hereditary optic neuropathy. Although DOA is caused by mutations in several genes, there are still many cases that have not been diagnosed or misdiagnosed. Herein, we present a large family of 11 patients with DOA. To identify potential pathogenic mutations, whole exome sequencing (WES) was performed on the proband, a 35-year-old woman. WES revealed a novel pathogenic mutation (c.524T>C, p.F175S) in the AFG3L2 intermembrane space domain, rather than in the ATPase domain, which is the hot mutation region associated with most of the previously reported DOA cases. Functional studies on skin fibroblasts generated from patients and HEK293T cells showed that the mutation may impair mitochondrial function and decrease the ability of AFG3L2 protein to enter the mitochondrial inner membrane. In addition, this novel mutation led to protein degradation and reduced the stability of the AFG3L2 protein, which appeared to be associated with the proteasome-ubiquitin pathway.
Collapse
|
33
|
Zhang Y, Zhang H, Yang Z, Zhang XH, Miao Q, Li M, Zhai TY, Zheng B, Wen JK. miR-155 down-regulation protects the heart from hypoxic damage by activating fructose metabolism in cardiac fibroblasts. J Adv Res 2022; 39:103-117. [PMID: 35777901 PMCID: PMC9263644 DOI: 10.1016/j.jare.2021.10.007] [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: 08/10/2021] [Revised: 10/15/2021] [Accepted: 10/17/2021] [Indexed: 12/09/2022] Open
Abstract
INTRODUCTION Hypoxia-inducible factor (HIF)1α has been shown to be activated and induces a glycolytic shift under hypoxic condition, however, little attention was paid to the role of HIF1α-actuated fructolysis in hypoxia-induced heart injury. OBJECTIVES In this study, we aim to explore the molecular mechanisms of miR-155-mediated fructose metabolism in hypoxic cardiac fibroblasts (CFs). METHODS Immunostaining, western blot and quantitative real-time reverse transcription PCR (qRT-PCR) were performed to detect the expression of glucose transporter 5 (GLUT5), ketohexokinase (KHK)-A and KHK-C in miR-155-/- and miR-155wt CFs under normoxia or hypoxia. A microarray analysis of circRNAs was performed to identify circHIF1α. Then CoIP, RIP and mass spectrometry analysis were performed and identified SKIV2L2 (MTR4) and transformer 2 alpha (TRA2A), a member of the transformer 2 homolog family. pAd-SKIV2L2 was administrated after coronary artery ligation to investigate whether SKIV2L2 can provide a protective effect on the infarcted heart. RESULTS When both miR-155-/- and miR-155wt CFs were exposed to hypoxia for 24 h, these two cells exhibited an increased glycolysis and decreased glycogen synthesis, and the expression of KHK-A and KHK-C, the central fructose-metabolizing enzyme, was upregulated. Mechanistically, miR-155 deletion in CFs enhanced SKIV2L2 expression and its interaction with TRA2A, which suppresses the alternative splicing of HIF1α pre-mRNA to form circHIF1α, and then decreased circHIF1α contributed to the activation of fructose metabolism through increasing the production of the KHK-C isoform. Finally, exogenous delivery of SKIV2L2 reduced myocardial damage in the infarcted heart. CONCLUSION In this study, we demonstrated that miR-155 deletion facilitates the activation of fructose metabolism in hypoxic CFs through regulating alternative splicing of HIF1α pre-mRNA and thus circHIF1ɑ formation.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Hong Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China; Department of Urology, Second Hospital of Hebei Medical University 050000, China
| | - Zhan Yang
- Department of Urology, Second Hospital of Hebei Medical University 050000, China
| | - Xin-Hua Zhang
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Qing Miao
- Department of Cardiovascular Medicine, Second Hospital of Hebei Medical University, 050000, China
| | - Min Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Tian-Ying Zhai
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China
| | - Bin Zheng
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China.
| | - Jin-Kun Wen
- Department of Biochemistry and Molecular Biology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, Hebei Medical University, Shijiazhuang 050017, China.
| |
Collapse
|
34
|
Uchikado Y, Ikeda Y, Ohishi M. Current Understanding of the Pivotal Role of Mitochondrial Dynamics in Cardiovascular Diseases and Senescence. Front Cardiovasc Med 2022; 9:905072. [PMID: 35665261 PMCID: PMC9157625 DOI: 10.3389/fcvm.2022.905072] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 04/14/2022] [Indexed: 12/11/2022] Open
Abstract
The heart is dependent on ATP production in mitochondria, which is closely associated with cardiovascular disease because of the oxidative stress produced by mitochondria. Mitochondria are highly dynamic organelles that constantly change their morphology to elongated (fusion) or small and spherical (fission). These mitochondrial dynamics are regulated by various small GTPases, Drp1, Fis1, Mitofusin, and Opa1. Mitochondrial fission and fusion are essential to maintain a balance between mitochondrial biogenesis and mitochondrial turnover. Recent studies have demonstrated that mitochondrial dynamics play a crucial role in the development of cardiovascular diseases and senescence. Disruptions in mitochondrial dynamics affect mitochondrial dysfunction and cardiomyocyte survival leading to cardiac ischemia/reperfusion injury, cardiomyopathy, and heart failure. Mitochondrial dynamics and reactive oxygen species production have been associated with endothelial dysfunction, which in turn causes the development of atherosclerosis, hypertension, and even pulmonary hypertension, including pulmonary arterial hypertension and chronic thromboembolic pulmonary hypertension. Here, we review the association between cardiovascular diseases and mitochondrial dynamics, which may represent a potential therapeutic target.
Collapse
Affiliation(s)
| | - Yoshiyuki Ikeda
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Sciences Kagoshima University, Kagoshima, Japan
| | | |
Collapse
|
35
|
Liu K, Li Y, Iqbal M, Tang Z, Zhang H. Thiram exposure in environment: A critical review on cytotoxicity. CHEMOSPHERE 2022; 295:133928. [PMID: 35149006 DOI: 10.1016/j.chemosphere.2022.133928] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/05/2022] [Accepted: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Thiram is used in large quantities in agriculture and may contaminate the environment by improper handling or storage in chemical plants and warehouses. A review of the literature has shown that thiram can affect different organs in animals and its toxic mechanisms can be elucidated in more detail at molecular level. We have summarized several impacts of thiram on animals: the effects of the perspectives of oxidative stress, mitochondrial damage, autophagy, apoptosis, and the IHH/PTHrP pathway on regulating abnormal skeletal development in particular tibial dyschondroplasia and kyphosis; angiogenesis inhibition was investigated from the perspective of angiogenesis factor inhibition, PI3K/AKT signaling pathway and CD147; the inhibition effect of thiram on fibroblasts and erythrocytes via the perspective of oxidative stress, mitochondrial damage and inhibition of growth factors in animal skin fibroblasts and erythrocytes; studied fertilized egg size, reduced fertility, neurodegeneration, and immune damage from the perspectives of CYP51 inhibition and dopamine-b-hydroxylase inhibition in the reproductive system, vitamin D deficiency in the nervous system, and inflammatory damage in the immune system; embryonic dysplasia in terms of thyroid hormone repression in animal embryonic development and repression of the SOX9a transcription factor. The elucidation of the mechanisms of toxicity of thiram on various organs of animals at molecular level will enable a more detailed understanding of the mechanisms of toxicity of thiram in animals and will facilitate the exploration of the treatment of thiram poisoning at molecular level.
Collapse
Affiliation(s)
- Kai Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Ying Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Mujahid Iqbal
- Department of Pathology, Cholistan University of Veterinary and Animal Sciences (CUVAS), Bahawalpur, 63100, Pakistan
| | - Zhaoxin Tang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China
| | - Hui Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, 510642, China.
| |
Collapse
|
36
|
Cui X, Tian Y, Zhao Y, Gao H, Yao D, Liu L, Li Y. miR-199b-5p-AKAP1-DRP1 pathway plays a key role in ox-LDL-induced mitochondrial fission and endothelial apoptosis. Curr Pharm Biotechnol 2022; 23:1612-1622. [PMID: 35331106 DOI: 10.2174/1389201023666220324123224] [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: 12/15/2021] [Revised: 01/13/2022] [Accepted: 02/01/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Atherosclerosis (AS) remains prevalent despite hyperlipidemia-lowering therapies. Although multiple functions of miR-199b-5p have been implicated in cancers, its role in endothelial apoptosis and AS remains unclear. This study aimed to examine the role of miR-199b-5p in mitochondrial dynamics and endothelial apoptosis. METHODS Human umbilical vein endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL) were subjected to other treatments, followed by a series analysis. We found that ox-LDL-treated HUVECs were associated with miR-199b-5p downregulation, increased reactive oxygen species level, reduced adenosine triphosphate (ATP) production, mitochondrial fission, and apoptosis, whereas enhanced miR-199b-5p expression or applied mitochondrial division inhibitor 1 (Mdivi-1) markedly reversed these changes. RESULTS Mechanistically, A-kinase anchoring protein 1 (AKAP1) was confirmed as a downstream target of miR-199b-5p by dual-luciferase activity reporter assay, AKAP1 overexpression reversed the anti-apoptotic effects of miR-199b-5p through the enhanced interaction of AKAP1 and dynamin protein 1 (DRP1) in ox-LDL-treated HUVECs. Moreover, miR-199b-5p downregulation, AKAP1 upregulation, and excessive mitochondrial fission were verified in human coronary AS endothelial tissues. CONCLUSION The miR-199b-5p-dependent regulation of AKAP1/DRP1 is required to inhibit hyperlipidemia-induced mitochondrial fission and endothelial injury and could be a promising therapeutic target for AS.
Collapse
Affiliation(s)
- Xiaolei Cui
- Emergency department of the second hospital of Hebei Medical University, China
| | - Yingping Tian
- Emergency department of the second hospital of Hebei Medical University, China
| | - Yapei Zhao
- Ultrasound department of the second hospital of Hebei Medical University, China
| | - Hengbo Gao
- Emergency department of the second hospital of Hebei Medical University, China
| | - Dongqi Yao
- Emergency department of the second hospital of Hebei Medical University, China
| | - Liang Liu
- Emergency department of the second hospital of Hebei Medical University, China
| | - Yongjun Li
- Department of Cardiology, the second hospital of Hebei Medical University, China
| |
Collapse
|
37
|
Li J, Wang Y, Wu T, Li S, Sun YN, Liu ZH. Baicalein suppresses high glucose-induced inflammation and apoptosis in trophoblasts by targeting the miRNA-17-5p-Mfn1/2-NF-κB pathway. Placenta 2022; 121:126-136. [DOI: 10.1016/j.placenta.2022.02.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 02/13/2022] [Accepted: 02/18/2022] [Indexed: 12/21/2022]
|
38
|
Ren H, Zhang YY, Li YL, Bai M, Yan QL, Huang XX, Cui W, Zhao H, Gu L, Liu Q, Yao GD, Song SJ. Semisynthesis and Non-Small-Cell Lung Cancer Cytotoxicity Evaluation of Germacrane-Type Sesquiterpene Lactones from Elephantopus scaber. JOURNAL OF NATURAL PRODUCTS 2022; 85:352-364. [PMID: 35090346 DOI: 10.1021/acs.jnatprod.1c00936] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Two series of germacrane-type sesquiterpene lactones were produced by semisynthetic modulation of scaberol C, which was prepared by a standard chemical transformation from an Elephantopus scaber extract. Their inhibition activities against non-small-cell lung cancer cells were screened, and preliminary structure-activity relationships were also established. Among them, monomeric analog 1u and dimeric analog 3d exhibited superior anti-non-small-cell lung cancer cytotoxic potencies with IC50 values of 4.3 and 0.7 μM against A549 cells, respectively, and were more active than cisplatin and the standard sesquiterpene lactones, parthenolide and scabertopin. Further studies revealed that compounds 1u and 3d cause G2/M phase arrest and induce apoptosis through the activation of mitochondrial pathways in A549 cells. Collectively, the results obtained suggest that compounds 1u and 3d are promising anti-non-small-cell lung cancer lead compounds.
Collapse
Affiliation(s)
- Hui Ren
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yang-Yang Zhang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Ya-Ling Li
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Ming Bai
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Qiu-Lin Yan
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xiao-Xiao Huang
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Wei Cui
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Hongwei Zhao
- Jilin Yizheng Pharmaceutical Group Co., Ltd., Siping 136001, Jilin Province, People's Republic of China
| | - Liwei Gu
- Institute of Chinese Materia Medica, Artemisinin Research Center, China Academy of Chinese Medical Sciences, Beijing 100700, People's Republic of China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
- Jilin Yizheng Pharmaceutical Group Co., Ltd., Siping 136001, Jilin Province, People's Republic of China
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| |
Collapse
|
39
|
Hu C, Zhang X, Teng T, Ma ZG, Tang QZ. Cellular Senescence in Cardiovascular Diseases: A Systematic Review. Aging Dis 2022; 13:103-128. [PMID: 35111365 PMCID: PMC8782554 DOI: 10.14336/ad.2021.0927] [Citation(s) in RCA: 72] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Aging is a prominent risk factor for cardiovascular diseases, which is the leading cause of death around the world. Recently, cellular senescence has received potential attention as a promising target in preventing cardiovascular diseases, including acute myocardial infarction, atherosclerosis, cardiac aging, pressure overload-induced hypertrophy, heart regeneration, hypertension, and abdominal aortic aneurysm. Here, we discuss the mechanisms underlying cellular senescence and describe the involvement of senescent cardiovascular cells (including cardiomyocytes, endothelial cells, vascular smooth muscle cells, fibroblasts/myofibroblasts and T cells) in age-related cardiovascular diseases. Then, we highlight the targets (SIRT1 and mTOR) that regulating cellular senescence in cardiovascular disorders. Furthermore, we review the evidence that senescent cells can exert both beneficial and detrimental implications in cardiovascular diseases on a context-dependent manner. Finally, we summarize the emerging pro-senescent or anti-senescent interventions and discuss their therapeutic potential in preventing cardiovascular diseases.
Collapse
Affiliation(s)
- Can Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Xin Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan 430060, China
| |
Collapse
|
40
|
Barba-Aliaga M, Alepuz P. Role of eIF5A in Mitochondrial Function. Int J Mol Sci 2022; 23:1284. [PMID: 35163207 PMCID: PMC8835957 DOI: 10.3390/ijms23031284] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/19/2022] [Accepted: 01/21/2022] [Indexed: 12/17/2022] Open
Abstract
The eukaryotic translation initiation factor 5A (eIF5A) is an evolutionarily conserved protein that binds ribosomes to facilitate the translation of peptide motifs with consecutive prolines or combinations of prolines with glycine and charged amino acids. It has also been linked to other molecular functions and cellular processes, such as nuclear mRNA export and mRNA decay, proliferation, differentiation, autophagy, and apoptosis. The growing interest in eIF5A relates to its association with the pathogenesis of several diseases, including cancer, viral infection, and diabetes. It has also been proposed as an anti-aging factor: its levels decay in aged cells, whereas increasing levels of active eIF5A result in the rejuvenation of the immune and vascular systems and improved brain cognition. Recent data have linked the role of eIF5A in some pathologies with its function in maintaining healthy mitochondria. The eukaryotic translation initiation factor 5A is upregulated under respiratory metabolism and its deficiency reduces oxygen consumption, ATP production, and the levels of several mitochondrial metabolic enzymes, as well as altering mitochondria dynamics. However, although all the accumulated data strongly link eIF5A to mitochondrial function, the precise molecular role and mechanisms involved are still unknown. In this review, we discuss the findings linking eIF5A and mitochondria, speculate about its role in regulating mitochondrial homeostasis, and highlight its potential as a target in diseases related to energy metabolism.
Collapse
Affiliation(s)
- Marina Barba-Aliaga
- Instituto de Biotecnología y Biomedicina (Biotecmed), Universitat de València, 46100 València, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, 46100 València, Spain
| | - Paula Alepuz
- Instituto de Biotecnología y Biomedicina (Biotecmed), Universitat de València, 46100 València, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, 46100 València, Spain
| |
Collapse
|
41
|
Huang C, Gao J, Wei T, Shen W. Angiotensin II-induced erythrocyte senescence contributes to oxidative stress. Rejuvenation Res 2021; 25:30-38. [PMID: 34969261 DOI: 10.1089/rej.2021.0054] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Oxidative stress may be an important cause of erythrocyte senescence. Angiotensin II (Ang II) has recently been shown to promote vascular cell senescence. However, its effects on erythrocytes remain unclear. This study aims to investigate the role of Ang II in regulating erythrocyte lifespan through oxidative stress. Experiments were performed in C57/BL6J mice infused with Ang II (1500 ng/kg per minute) or saline for 7 days. Following Ang II infusion, we found that Ang II increased erythrocyte number, hemoglobin and red blood cell distribution width (RDW). These differences were accompanied by a decrease in glutathione (GSH) and an increase in malondialdehyde (MDA) concentration. In vitro, after 24 hours of Ang II treatment, erythrocytes showed reduced surface expression of CD47 and increased phosphatidylserine exposure. In parallel, Ang II reduced the levels of antioxidant enzymes, including Cu/ZnSOD, catalase, and peroxidase 2 (PRDX2). These effects were reversed by the addition of the antioxidant N-acetyl-L-cysteine or the Ang II type 1 receptor (AT1) blocker losartan. In addition, Ang II treatment increased pro-inflammatory oxylipin, including hydroxyeicosatetraenoic acids (HETEs) and dihydroxyoctadecenoic acids (DiHOMEs) in the erythrocyte membranes. Collectively, Ang II induced erythrocyte senescence and susceptibility to eryptosis, partially due to enhanced oxidative stress.
Collapse
Affiliation(s)
- Chenglin Huang
- Shanghai Institute of Hypertension, 194034, Shanghai, Shanghai, China;
| | - Jing Gao
- Shanghai Institute of Hypertension, 194034, Shanghai, China;
| | - Tong Wei
- Shanghai Institute of Hypertension, 194034, Shanghai, China;
| | - Weili Shen
- Shanghai Institute of Hypertension, 194034, 197,2nd Ruijin road, Shanghai, China, 200025;
| |
Collapse
|
42
|
Tauc M, Cougnon M, Carcy R, Melis N, Hauet T, Pellerin L, Blondeau N, Pisani DF. The eukaryotic initiation factor 5A (eIF5A1), the molecule, mechanisms and recent insights into the pathophysiological roles. Cell Biosci 2021; 11:219. [PMID: 34952646 PMCID: PMC8705083 DOI: 10.1186/s13578-021-00733-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/14/2021] [Indexed: 11/29/2022] Open
Abstract
Since the demonstration of its involvement in cell proliferation, the eukaryotic initiation factor 5A (eIF5A) has been studied principally in relation to the development and progression of cancers in which the isoform A2 is mainly expressed. However, an increasing number of studies report that the isoform A1, which is ubiquitously expressed in normal cells, exhibits novel molecular features that reveal its new relationships between cellular functions and organ homeostasis. At a first glance, eIF5A can be regarded, among other things, as a factor implicated in the initiation of translation. Nevertheless, at least three specificities: (1) its extreme conservation between species, including plants, throughout evolution, (2) its very special and unique post-translational modification through the activating-hypusination process, and finally (3) its close relationship with the polyamine pathway, suggest that the role of eIF5A in living beings remains to be uncovered. In fact, and beyond its involvement in facilitating the translation of proteins containing polyproline residues, eIF5A is implicated in various physiological processes including ischemic tolerance, metabolic adaptation, aging, development, and immune cell differentiation. These newly discovered physiological properties open up huge opportunities in the clinic for pathologies such as, for example, the ones in which the oxygen supply is disrupted. In this latter case, organ transplantation, myocardial infarction or stroke are concerned, and the current literature defines eIF5A as a new drug target with a high level of potential benefit for patients with these diseases or injuries. Moreover, the recent use of genomic and transcriptomic association along with metadata studies also revealed the implication of eIF5A in genetic diseases. Thus, this review provides an overview of eIF5A from its molecular mechanism of action to its physiological roles and the clinical possibilities that have been recently reported in the literature.
Collapse
Affiliation(s)
- Michel Tauc
- LP2M, CNRS, Université Côte d'Azur, Nice, France. .,Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France. .,Laboratoire de Physiomédecine Moléculaire, UMR7370, Faculté de Médecine, CNRS, Université Côte d'Azur, 28 Avenue de Valombrose, 06107, Nice Cedex, France.
| | - Marc Cougnon
- LP2M, CNRS, Université Côte d'Azur, Nice, France.,Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| | - Romain Carcy
- Service de Réanimation Polyvalente et Service de Réanimation des Urgences Vitales, CHU Nice, Hôpital Pasteur 2, Nice, France
| | - Nicolas Melis
- Laboratory of Cellular and Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Thierry Hauet
- INSERM, IRTOMIT, CHU de Poitiers, Université de Poitiers, La Milétrie, Poitiers, France
| | - Luc Pellerin
- INSERM, IRTOMIT, CHU de Poitiers, Université de Poitiers, La Milétrie, Poitiers, France
| | - Nicolas Blondeau
- Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France.,IPMC, CNRS, Université Côte d'Azur, Valbonne, France
| | - Didier F Pisani
- LP2M, CNRS, Université Côte d'Azur, Nice, France.,Laboratories of Excellence Ion Channel Science and Therapeutics, Nice, France
| |
Collapse
|
43
|
Luan Y, Ren KD, Luan Y, Chen X, Yang Y. Mitochondrial Dynamics: Pathogenesis and Therapeutic Targets of Vascular Diseases. Front Cardiovasc Med 2021; 8:770574. [PMID: 34938787 PMCID: PMC8685340 DOI: 10.3389/fcvm.2021.770574] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular diseases, particularly atherosclerosis, are associated with high morbidity and mortality. Endothelial cell (EC) or vascular smooth muscle cell (VSMC) dysfunction leads to blood vessel abnormalities, which cause a series of vascular diseases. The mitochondria are the core sites of cell energy metabolism and function in blood vessel development and vascular disease pathogenesis. Mitochondrial dynamics, including fusion and fission, affect a variety of physiological or pathological processes. Multiple studies have confirmed the influence of mitochondrial dynamics on vascular diseases. This review discusses the regulatory mechanisms of mitochondrial dynamics, the key proteins that mediate mitochondrial fusion and fission, and their potential effects on ECs and VSMCs. We demonstrated the possibility of mitochondrial dynamics as a potential target for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Yi Luan
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai-Di Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Ying Luan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xing Chen
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
44
|
Mangiferin Inhibits PDGF-BB-Induced Proliferation and Migration of Rat Vascular Smooth Muscle Cells and Alleviates Neointimal Formation in Mice through the AMPK/Drp1 Axis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3119953. [PMID: 34900084 PMCID: PMC8664531 DOI: 10.1155/2021/3119953] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 10/19/2021] [Indexed: 01/15/2023]
Abstract
Mangiferin is a naturally occurring xanthone C-glycoside that is widely found in various plants. Previous studies have reported that mangiferin inhibits tumor cell proliferation and migration. Excessive proliferation and migration of vascular smooth muscle cells (SMCs) is associated with neointimal hyperplasia in coronary arteries. However, the role and mechanism of mangiferin action in neointimal hyperplasia is still unknown. In this study, a mouse carotid artery ligation model was established, and primary rat smooth muscle cells were isolated and used for mechanistic assays. We found that mangiferin alleviated neointimal hyperplasia, inhibited proliferation and migration of SMCs, and promoted platelets derive growth factors-BB- (PDGF-BB-) induced contractile phenotype in SMCs. Moreover, mangiferin attenuated neointimal formation by inhibiting mitochondrial fission through the AMPK/Drp1 signaling pathway. These findings suggest that mangiferin has the potential to maintain vascular homeostasis and inhibit neointimal hyperplasia.
Collapse
|
45
|
Wang Y, Gan Z, Lu H, Liu Z, Shang P, Zhang J, Yin W, Chu H, Yuan R, Ye Y, Chen P, Rong M. Impact of High-Altitude Hypoxia on Early Osseointegration With Bioactive Titanium. Front Physiol 2021; 12:689807. [PMID: 35035356 PMCID: PMC8753411 DOI: 10.3389/fphys.2021.689807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 10/22/2021] [Indexed: 01/03/2023] Open
Abstract
Nowadays, the bone osseointegration in different environments is comparable, but the mechanism is unclear. This study aimed to investigate the osseointegration of different bioactive titanium surfaces under normoxic or high-altitude hypoxic environments. Titanium implants were subjected to one of two surface treatments: (1) sanding, blasting, and acid etching to obtain a rough surface, or (2) extensive polishing to obtain a smooth surface. Changes in the morphology, proliferation, and protein expression of osteoblasts on the rough and smooth surfaces were examined, and bone formation was studied through western blotting and animal-based experiments. Our findings found that a hypoxic environment and rough titanium implant surface promoted the osteogenic differentiation of osteoblasts and activated the JAK1/STAT1/HIF-1α pathway in vitro. The animal study revealed that following implant insertion in tibia of rabbit, bone repair at high altitudes was slower than that at low altitudes (i.e., in plains) after 2weeks; however, bone formation did not differ significantly after 4weeks. The results of our study showed that: (1) The altitude hypoxia environment would affect the early osseointegration of titanium implants while titanium implants with rough surfaces can mitigate the effects of this hypoxic environment on osseointegration, (2) the mechanism may be related to the activation of JAK1/STAT1/HIF-1α pathway, and (3) our results suggest the osteogenesis of titanium implants, such as oral implants, is closely related to the oxygen environment. Clinical doctors, especially dentists, should pay attention to the influence of hypoxia on early osseointegration in patients with high altitude. For example, it is better to choose an implant system with rough implant surface in the oral cavity of patients with tooth loss at high altitude.
Collapse
Affiliation(s)
- Yarong Wang
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Zekun Gan
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Haibin Lu
- Department of Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Ziyi Liu
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Peng Shang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Jian Zhang
- College of Animal Science, Tibet Agriculture and Animal Husbandry University, Linzhi, China
| | - Wuwei Yin
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Hongxing Chu
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | | | - Yingxin Ye
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Pei Chen
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
- Pei Chen,
| | - Mingdeng Rong
- Department of Periodontology and Implantology, Stomatological Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Mingdeng Rong,
| |
Collapse
|
46
|
Nguyen DDN, Zain SM, Kamarulzaman MH, Low TY, Chilian WM, Pan Y, Ting KN, Hamid A, Abdul Kadir A, Pung YF. Intracellular and exosomal microRNAome profiling of human vascular smooth muscle cells during replicative senescence. Am J Physiol Heart Circ Physiol 2021; 321:H770-H783. [PMID: 34506226 DOI: 10.1152/ajpheart.00058.2021] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular aging is highly associated with cardiovascular morbidity and mortality. Although the senescence of vascular smooth muscle cells (VSMCs) has been well established as a major contributor to vascular aging, intracellular and exosomal microRNA (miRNA) signaling pathways in senescent VSMCs have not been fully elucidated. This study aimed to identify the differential expression of intracellular and exosomal miRNA in human VSMCs (hVSMCs) during replicative senescence. To achieve this aim, intracellular and exosomal miRNAs were isolated from hVSMCs and subsequently subjected to whole genome small RNA next-generation sequencing, bioinformatics analyses, and qPCR validation. Three significant findings were obtained. First, senescent hVSMC-derived exosomes tended to cluster together during replicative senescence and the molecular weight of the exosomal protein tumor susceptibility gene 101 (TSG-101) increased relative to the intracellular TSG-101, suggesting potential posttranslational modifications of exosomal TSG-101. Second, there was a significant decrease in both intracellular and exosomal hsa-miR-155-5p expression [n = 3, false discovery rate (FDR) < 0.05], potentially being a cell type-specific biomarker of hVSMCs during replicative senescence. Importantly, hsa-miR-155-5p was found to associate with cell-cycle arrest and elevated oxidative stress. Lastly, miRNAs from the intracellular pool, that is, hsa-miR-664a-3p, hsa-miR-664a-5p, hsa-miR-664b-3p, hsa-miR-4485-3p, hsa-miR-10527-5p, and hsa-miR-12136, and that from the exosomal pool, that is, hsa-miR-7704, were upregulated in hVSMCs during replicative senescence (n = 3, FDR < 0.05). Interestingly, these novel upregulated miRNAs were not functionally well annotated in hVSMCs to date. In conclusion, hVSMC-specific miRNA expression profiles during replicative senescence potentially provide valuable insights into the signaling pathways leading to vascular aging.NEW & NOTEWORTHY This is the first study on intracellular and exosomal miRNA profiling on human vascular smooth muscle cells during replicative senescence. Specific dysregulated sets of miRNAs were identified from human vascular smooth muscle cells. Hsa-miR-155-5p was significantly downregulated in both intracellular and exosomal hVSMCs, suggesting its crucial role in cellular senescence. Hsa-miR-155-5p might be the mediator in linking cellular senescence to vascular aging and atherosclerosis.
Collapse
Affiliation(s)
- Diem Duong Ngoc Nguyen
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Shamsul Mohd Zain
- The Pharmacogenomics Laboratory, Department of Pharmacology, University of Malaya, Kuala Lumpur, Malaysia
| | | | - Teck Yew Low
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - William M Chilian
- Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, Ohio
| | - Yan Pan
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Kang Nee Ting
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Aini Hamid
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| | - Arifah Abdul Kadir
- Department of Veterinary Preclinical Sciences, Faculty of Veterinary Medicine, University Putra Malaysia, Selangor, Malaysia
| | - Yuh-Fen Pung
- Division of Biomedical Science, School of Pharmacy, University of Nottingham Malaysia, Selangor, Malaysia
| |
Collapse
|
47
|
Weng G, Gu M, Zhang Y, Zhao G, Gu Y. LINC01123 promotes cell proliferation and migration via regulating miR-1277-5p/KLF5 axis in ox-LDL-induced vascular smooth muscle cells. J Mol Histol 2021; 52:943-953. [PMID: 34403009 DOI: 10.1007/s10735-021-10010-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 08/09/2021] [Indexed: 11/25/2022]
Abstract
The pathophysiological mechanism of carotid atherosclerosis (CAS) involves endothelial cell dysfunction, vascular smooth muscle cells (VSMCs), and macrophage activation, which ultimately leads to fibrosis of the vessel wall. lncRNA works weightily in the formation of CAS, but the function and mechanism of lncRNA LINC01123 in stable plaque formation are still equivocal. We collected blood samples from 35 CAS patients as well as 33 healthy volunteers. VSMCs treated with oxidized low-density lipoprotein (ox-LDL) were utilized as the CAS cell models. We applied qRT-PCR for detecting LINC01123, miR-1277-5p and KLF5 mRNA expression, CCK-8 method and BrdU test for determining cell proliferation, Transwell test for measuring cell migration, as well as Western blot for assaying KLF5 protein expression. Dual-luciferase reporter experiment was adopted for assessing the interaction between LINC01123 and miR-1277-5p, as well as KLF5 and miR-1277-5p. LINC01123 and KLF5 expression were dramatically up-regulated, while miR-1277-5p expression was down-regulated in CAS patients and ox-LDL-induced CAS cell models. Overexpressed LINC01123 notedly promoted VSMCs migration and proliferation. LINC01123 knockdown repressed cell proliferation and migration. Also, LINC01123 targeted miR-1277-5p and down-regulated its expression, while miR-1277-5p could negatively regulate KLF5 expression. LINC01123 is highly expressed in CAS patients, and promotes cell proliferation and migration via regulating miR-1277-5p/KLF5 axis in ox-LDL-induced VSMCs. It might be involved in the fibrous plaque formation.
Collapse
Affiliation(s)
- Guohu Weng
- Department of Encephalopathy, Hainan Provincial Hospital of Chinese Medicine, 47# Heping North Road, Haikou, 570203, Hainan, People's Republic of China
| | - Minhua Gu
- Department of Cardiology, Hainan Provincial Hospital of Chinese Medicine, Haikou, 570203, Hainan, People's Republic of China
| | - Yifan Zhang
- Department of Encephalopathy, Hainan Provincial Hospital of Chinese Medicine, 47# Heping North Road, Haikou, 570203, Hainan, People's Republic of China
| | - Guangfeng Zhao
- Department of Encephalopathy, Hainan Provincial Hospital of Chinese Medicine, 47# Heping North Road, Haikou, 570203, Hainan, People's Republic of China
| | - Yong Gu
- Department of Encephalopathy, Hainan Provincial Hospital of Chinese Medicine, 47# Heping North Road, Haikou, 570203, Hainan, People's Republic of China.
| |
Collapse
|
48
|
Pereira KD, Tamborlin L, de Lima TI, Consonni SR, Silveira LR, Luchessi AD. Alternative human eIF5A protein isoform plays a critical role in mitochondria. J Cell Biochem 2021; 122:549-561. [PMID: 33459432 DOI: 10.1002/jcb.29884] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/24/2020] [Accepted: 12/21/2020] [Indexed: 02/06/2023]
Abstract
The eukaryotic translation initiation factor 5A (eIF5A) is the only known protein containing the amino acid residue hypusine, essential for its activity. Hypusine residue is produced by a posttranslational modification involving deoxyhypusine synthetase and deoxyhypusine hydroxylase. Herein, we aimed to describe the role of the alternative human isoform A on mitochondrial processes. Isoform A depletion modulates oxidative metabolism in association with the downregulation of mitochondrial biogenesis-related genes. Through positive feedback, it increases cell respiration leading to highly reactive oxygen species production, which impacts mitochondrial bioenergetics. These metabolic changes compromise mitochondrial morphology, increasing its electron density and fission, observed by transmission electron microscopy. This set of changes leads the cells to apoptosis, evidenced by increased DNA fragmentation and proapoptotic BAK protein content increase. Thus, we show that the alternative eIF5A isoform A is crucial for energy metabolism controlled by mitochondria and cellular survival.
Collapse
Affiliation(s)
- Karina D Pereira
- Laboratory of Biotechnology, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Letícia Tamborlin
- Laboratory of Biotechnology, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| | - Tanes I de Lima
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Silvio R Consonni
- Laboratory of Cytochemistry and Immunocytochemistry, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, São Paulo, Brazil
| | - Leonardo R Silveira
- Department of Structural and Functional Biology, Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Augusto D Luchessi
- Laboratory of Biotechnology, School of Applied Sciences, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- Institute of Biosciences, São Paulo State University (UNESP), Rio Claro, São Paulo, Brazil
| |
Collapse
|
49
|
Gao L, Yang X, Li Y, Wang Z, Wang S, Tan S, Chen A, Cao P, Shao J, Zhang Z, Zhang F, Zheng S. Curcumol inhibits KLF5-dependent angiogenesis by blocking the ROS/ERK signaling in liver sinusoidal endothelial cells. Life Sci 2021; 264:118696. [PMID: 33157090 DOI: 10.1016/j.lfs.2020.118696] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/18/2020] [Accepted: 10/28/2020] [Indexed: 12/12/2022]
Abstract
AIMS Liver fibrosis is a difficult problem in the medical field. We previously reported that curcumol, a bioactive substance, may inhibit the pathological angiogenesis of liver sinusoidal endothelial cells (LSECs) and play a good anti-hepatic fibrosis effect. However, the mechanism of curcumol inhibiting angiogenesis in LSEC needs to be further clarified. Here, we focus on how curcumol inhibits LSEC angiogenesis in liver fibrosis. MATERIALS AND METHODS Primary rat LSECs were cultured in vitro, and various molecular experiments including real-time PCR, western blot, immunofluorescence, tube formation assay and transwell migration assay were used to clarify the potential mechanism of curcumol. Carbon tetrachloride (CCl4) was applied to create a mouse liver fibrosis model. Blood and livers were taken to elucidate the efficacy of curcumol in vivo. KEY FINDINGS We found that curcumol could effectively inhibit LSEC angiogenesis in vitro. Interestingly, this process may depend on curcumol's inhibition of the expression of transcription factor KLF5. Mice experiment also showed that curcumol could alleviate chronic liver injury by reducing KLF5 expression. In addition, we suggested that curcumol could reduce the production of mitochondrial ROS and improve mitochondrial morphology in LSEC. More importantly, we proved that curcumol could suppress KLF5-mediated LSEC angiogenesis by inhibiting ROS/ERK signaling. SIGNIFICANCE We suggested that transcription factor KLF5 could be considered as a new target molecule of curcumol in improving liver fibrosis, and pointed out that curcumol targeted ROS/ERK-mediated KLF5 expression could inhibit LSEC angiogenesis. This provided a new theoretical basis for curcumol to ameliorate liver fibrosis.
Collapse
Affiliation(s)
- Liyuan Gao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiang Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Li
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhenyi Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shijun Wang
- Shandong Co-innovation Center of TCM Formula, College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shanzhong Tan
- Department of Integrated TCM and Western Medicine, Nanjing Hospital Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Anping Chen
- Department of Pathology, School of Medicine, Saint Louis University, St Louis, USA
| | - Peng Cao
- Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiangjuan Shao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zili Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Feng Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| | - Shizhong Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Nanjing University of Chinese Medicine, Nanjing, China; Jiangsu Key Laboratory of Therapeutic Material of Chinese Medicine, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
50
|
Barba-Aliaga M, Villarroel-Vicente C, Stanciu A, Corman A, Martínez-Pastor MT, Alepuz P. Yeast Translation Elongation Factor eIF5A Expression Is Regulated by Nutrient Availability through Different Signalling Pathways. Int J Mol Sci 2020; 22:E219. [PMID: 33379337 PMCID: PMC7794953 DOI: 10.3390/ijms22010219] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/17/2020] [Accepted: 12/24/2020] [Indexed: 12/15/2022] Open
Abstract
Translation elongation factor eIF5A binds to ribosomes to promote peptide bonds between problematic amino acids for the reaction like prolines. eIF5A is highly conserved and essential in eukaryotes, which usually contain two similar but differentially expressed paralogue genes. The human eIF5A-1 isoform is abundant and implicated in some cancer types; the eIF5A-2 isoform is absent in most cells but becomes overexpressed in many metastatic cancers. Several reports have connected eIF5A and mitochondria because it co-purifies with the organelle or its inhibition reduces respiration and mitochondrial enzyme levels. However, the mechanisms of eIF5A mitochondrial function, and whether eIF5A expression is regulated by the mitochondrial metabolism, are unknown. We analysed the expression of yeast eIF5A isoforms Tif51A and Tif51B under several metabolic conditions and in mutants. The depletion of Tif51A, but not Tif51B, compromised yeast growth under respiration and reduced oxygen consumption. Tif51A expression followed dual positive regulation: by high glucose through TORC1 signalling, like other translation factors, to promote growth and by low glucose or non-fermentative carbon sources through Snf1 and heme-dependent transcription factor Hap1 to promote respiration. Upon iron depletion, Tif51A was down-regulated and Tif51B up-regulated. Both were Hap1-dependent. Our results demonstrate eIF5A expression regulation by cellular metabolic status.
Collapse
Affiliation(s)
- Marina Barba-Aliaga
- Instituto Biotecmed, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain; (M.B.-A.); (C.V.-V.); (A.S.); (A.C.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain;
| | - Carlos Villarroel-Vicente
- Instituto Biotecmed, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain; (M.B.-A.); (C.V.-V.); (A.S.); (A.C.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain;
| | - Alice Stanciu
- Instituto Biotecmed, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain; (M.B.-A.); (C.V.-V.); (A.S.); (A.C.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain;
| | - Alba Corman
- Instituto Biotecmed, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain; (M.B.-A.); (C.V.-V.); (A.S.); (A.C.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain;
| | - María Teresa Martínez-Pastor
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain;
| | - Paula Alepuz
- Instituto Biotecmed, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain; (M.B.-A.); (C.V.-V.); (A.S.); (A.C.)
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universitat de València, C/Dr. Moliner 50, E46100 Burjassot, Spain;
| |
Collapse
|