1
|
Wei C, Liu X, Miao Z, Zhang H, Wang Y, Qi G. TWEAK/Fn14 axis may promote vascular smooth muscle cell senescence via p38 signaling pathway: preliminary evidence. Future Sci OA 2025; 11:2455906. [PMID: 39840833 PMCID: PMC11756581 DOI: 10.1080/20565623.2025.2455906] [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: 01/03/2024] [Accepted: 12/12/2024] [Indexed: 01/23/2025] Open
Abstract
AIM The primary objective of this study is to investigate the impact of tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its functional receptor, fibroblast growth factor-inducible 14 (Fn14), on the process of vascular smooth muscle cell (VSMC) senescence. METHODS Rat arterial VSMCs were cultured with angiotensin II to establish a model of premature senescence. The effects of TWEAK and Fn14 on senescent VSMCs were evaluated. Additionally, the role of p38 phosphorylation pathway in the effect of TWEAK on VSMCs senescence was assessed. RESULTS Expressions of TWEAK and Fn14 were significantly elevated in senescent VSMCs. TWEAK activated the p38 phosphorylation pathway and promoted the SA-β-gal staining and P53 expression. CONCLUSION These preliminary findings suggest that the TWEAK/Fn14 axis may play a crucial role in promoting VSMC senescence.
Collapse
Affiliation(s)
- Chunyang Wei
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiaoying Liu
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Zhuang Miao
- Department of Anesthesiology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Hua Zhang
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yanfu Wang
- Department of Gerontology, the First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Guoxian Qi
- Department of Gerontology, the First Affiliated Hospital, China Medical University, Shenyang, China
| |
Collapse
|
2
|
De Bartolo A, Angelone T, Rocca C. Elucidating emerging signaling pathways driving endothelial dysfunction in cardiovascular aging. Vascul Pharmacol 2025; 158:107462. [PMID: 39805379 DOI: 10.1016/j.vph.2025.107462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/24/2024] [Accepted: 01/03/2025] [Indexed: 01/16/2025]
Abstract
The risk for developing cardiovascular diseases dramatically increases in older individuals, and aging vasculature plays a crucial role in determining their morbidity and mortality. Aging disrupts endothelial balance between vasodilators and vasoconstrictors, impairing function and promoting pathological vascular remodeling. In this Review, we discuss the impact of key and emerging molecular pathways that transduce aberrant inflammatory signals (i.e., chronic low-grade inflammation-inflammaging), oxidative stress, and mitochondrial dysfunction in aging vascular compartment. We focus on the interplay between these events, which contribute to generating a vicious cycle driving the progressive alterations in vascular structure and function during cardiovascular aging. We also discuss the primary role of senescent endothelial cells and vascular smooth muscle cells, and the potential link between vascular and myeloid cells, in impairing plaque stability and promoting the progression of atherosclerosis. The aim of this summary is to provide potential novel insights into targeting these processes for therapeutic benefit.
Collapse
Affiliation(s)
- Anna De Bartolo
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy
| | - Tommaso Angelone
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, Department of Biology, E. and E. S. (DiBEST), University of Calabria, Arcavacata di Rende, Cosenza, Italy; National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
3
|
Yousef A, Fang L, Heidari M, Kranrod J, Seubert JM. The role of CYP-sEH derived lipid mediators in regulating mitochondrial biology and cellular senescence: implications for the aging heart. Front Pharmacol 2024; 15:1486717. [PMID: 39703395 PMCID: PMC11655241 DOI: 10.3389/fphar.2024.1486717] [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: 08/26/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
Cellular senescence is a condition characterized by stable, irreversible cell cycle arrest linked to the aging process. The accumulation of senescent cells in the cardiac muscle can contribute to various cardiovascular diseases (CVD). Telomere shortening, epigenetic modifications, DNA damage, mitochondrial dysfunction, and oxidative stress are known contributors to the onset of cellular senescence in the heart. The link between mitochondrial processes and cellular senescence contributed to the age-related decline in cardiac function. These include changes in mitochondrial functions and behaviours that arise from various factors, including impaired dynamics, dysregulated biogenesis, mitophagy, mitochondrial DNA (mtDNA), reduced respiratory capacity, and mitochondrial structural changes. Thus, regulation of mitochondrial biology has a role in cellular senescence and cardiac function in aging hearts. Targeting senescent cells may provide a novel therapeutic approach for treating and preventing CVD associated with aging. CYP epoxygenases metabolize N-3 and N-6 polyunsaturated fatty acids (PUFA) into epoxylipids that are readily hydrolyzed to diol products by soluble epoxide hydrolase (sEH). Increasing epoxylipids levels or inhibition of sEH has demonstrated protective effects in the aging heart. Evidence suggests they may play a role in cellular senescence by regulating mitochondria, thus reducing adverse effects of aging in the heart. In this review, we discuss how mitochondria induce cellular senescence and how epoxylipids affect the senescence process in the aged heart.
Collapse
Affiliation(s)
- Ala Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Liye Fang
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mobina Heidari
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Joshua Kranrod
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
4
|
Fakfum P, Chuljerm H, Parklak W, Roytrakul S, Phaonakrop N, Lerttrakarnnon P, Kulprachakarn K. Plasma Proteomics of Type 2 Diabetes, Hypertension, and Co-Existing Diabetes/Hypertension in Thai Adults. Life (Basel) 2024; 14:1269. [PMID: 39459569 PMCID: PMC11509282 DOI: 10.3390/life14101269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/31/2024] [Accepted: 10/03/2024] [Indexed: 10/28/2024] Open
Abstract
The study explored proteomics to better understand the relationship between type 2 diabetes (T2DM) and hypertension (HT) in Thai adults, using shotgun proteomics and bioinformatics analysis. Plasma samples were taken from 61 subjects: 14 healthy subjects (mean age = 40.85 ± 7.12), 13 with T2DM (mean age = 57.38 ± 6.03), 16 with HT (mean age = 66.87 ± 10.09), and 18 with coexisting T2DM/HT (mean age = 58.22 ± 10.65). Proteins were identified using liquid chromatography-tandem mass spectrometry (LC-MS/MS). Protein-protein interactions were analyzed using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) version 11.5. We identified six unique proteins in T2DM patients, including translationally controlled 1 (TPT1) and nibrin (NBN), which are associated with the DNA damage response. In HT patients, seven unique proteins were identified, among them long-chain fatty acid-CoA ligase (ASCL), which functions in the stimulation of triacylglycerol and cholesterol synthesis, and NADPH oxidase activator 1 (NOXA1), which is involved in high blood pressure via angiotensin II-induced reactive oxygen species (ROS)-generating systems. In coexisting T2DM/HT patients, six unique proteins were identified, of which two-microtubule-associated protein 1A (MAP1A)-might be involved in dementia via RhoB-p53 and diacylglycerol kinase beta (DGKB), associated with lipid metabolism. This study identified new candidate proteins that are possibly involved in the pathology of these diseases.
Collapse
Affiliation(s)
- Puriwat Fakfum
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| | - Hataichanok Chuljerm
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| | - Wason Parklak
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| | - Sittiruk Roytrakul
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Narumon Phaonakrop
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency, Pathum Thani 12120, Thailand; (S.R.); (N.P.)
| | - Peerasak Lerttrakarnnon
- Aging and Aging Palliative Care Research Cluster, Department of Family Medicine, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Kanokwan Kulprachakarn
- Research Institute for Health Sciences, Chiang Mai University, Chiang Mai 50200, Thailand; (P.F.); (H.C.); (W.P.)
| |
Collapse
|
5
|
Guan Y, Zhang Y, Chen L, Ren Y, Nie H, Ji T, Yan J, Zhang C, Ruan L. Effect of low-dose terazosin on arterial stiffness improvement: A pilot study. J Cell Mol Med 2024; 28:e18547. [PMID: 39044238 PMCID: PMC11265993 DOI: 10.1111/jcmm.18547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 06/26/2024] [Accepted: 07/12/2024] [Indexed: 07/25/2024] Open
Abstract
Arterial stiffness, a prominent hallmark of ageing arteries, is a predictor of all-cause mortality. Strategies for promoting healthy vascular ageing are encouraged. Here we conducted a pilot study to evaluate the potential effects of low-dose Terazosin on arterial stiffness. We enrolled patients aged over 40 with elevated arterial stiffness, defined as a brachial-ankle pulse wave velocity (baPWV) ≥1400 cm/s, who were administered Terazosin (0.5 and 1.0 mg/day) from December 2020 to June 2023. Treatment responses were assessed every 3 months. Linear regression analysis was used to characterise the improvement. We matched cases who took Terazosin for 1 year with Terazosin-free controls using propensity score matching (PSM). Our findings demonstrate that Terazosin administration significantly affected arterial stiffness. (1) Arterial stiffness significantly improved (at least a 5% reduction in baPWV) in 50.0% of patients at 3 months, 48.6% at 6 months, 59.3% at 9 months, and 54.4% at 12 months, respectively. (2) Those with higher baseline baPWV and hypertension exhibited a significantly reduced risk of non-response. (3) Terazosin was associated with a reduction of baPWV at 1-year follow-up (linear regression: β = -165.16, p < 0.001). This pilot study offers valuable insights into the potential significance of Terazosin in improving arterial stiffness and paves the way for future randomised clinical trials in combating vascular ageing.
Collapse
Affiliation(s)
- Yuqi Guan
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yucong Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Liangkai Chen
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Ministry of Education Key Lab of Environment and Health, School of Public Health, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Yazhi Ren
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Hao Nie
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Tianyi Ji
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jinhua Yan
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Cuntai Zhang
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lei Ruan
- Department of Geriatrics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| |
Collapse
|
6
|
Olopade EO, Morakinyo AE, Alao JO, Oyedepo TA. Effects of n-hexane fraction of Piper guineense seed extract on N ω-nitro-L-arginine methyl ester hydrochloride-induced hypertension in rats. Cell Biochem Funct 2024; 42:e4095. [PMID: 39004810 DOI: 10.1002/cbf.4095] [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: 05/07/2024] [Revised: 06/14/2024] [Accepted: 07/04/2024] [Indexed: 07/16/2024]
Abstract
This study aimed to investigate the effects of the n-hexane fraction of the ethanolic seed extract of PG (NFESEPG) on hypertension induced by Nω-nitro-L-arginine methyl ester (L-NAME) in rats. Specifically, the study examined the impact of NFESEPG on blood pressure, oxidative stress markers, NO concentration, angiotensin-converting enzyme (ACE) and arginase activities, and cardiac biomarkers in hypertensive rats. The study involved collecting, identifying, and processing the PG plant to obtain the ethanolic seed extract. The extract was then partitioned with solvents to isolate the n-hexane fraction. Hypertension was induced in rats by oral administration of L-NAME for 10 days, while concurrent treatment with NFESEPG at two doses (200 and 400 mg/kg/day) was administered orally. Blood pressure was measured using a noninvasive tail-cuff method, and various biochemical parameters were assessed. Treatment with both doses of NFESEPG significantly reduced systolic and diastolic blood pressure in L-NAME-induced hypertensive rats. Additionally, NFESEPG administration increased NO concentration and decreased ACE and arginase activities, malondialdehyde (MDA) levels, and cardiac biomarkers in hypertensive rats. The findings indicate that NFESEPG effectively lowered blood pressure in hypertensive rats induced by L-NAME, potentially through mechanisms involving the modulation of oxidative stress, NO bioavailability, and cardiac biomarkers. These results suggest the therapeutic potential of NFESEPG in managing hypertension and related cardiovascular complications.
Collapse
Affiliation(s)
| | | | - Jude Oluwapelumi Alao
- School of Public Health and Interdisciplinary Studies, Auckland University of Technology, Auckland, New Zealand
| | | |
Collapse
|
7
|
Ke P, Xie J, Xu T, Chen M, Guo Y, Wang Y, Qiu H, Wu D, Zeng Z, Chen S, Bao X. Identification of a venetoclax-resistance prognostic signature base on 6-senescence genes and its clinical significance for acute myeloid leukemia. Front Oncol 2023; 13:1302356. [PMID: 38098504 PMCID: PMC10720639 DOI: 10.3389/fonc.2023.1302356] [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: 09/26/2023] [Accepted: 11/14/2023] [Indexed: 12/17/2023] Open
Abstract
Background Satisfactory responses can be obtained for acute myeloid leukemia (AML) treated by Venetoclax (VEN)-based therapy. However, there are still quite a few AML patients (AMLs) resistant to VEN, and it is critical to understand whether VEN-resistance is regulated by senescence. Methods Here, we established and validated a signature for predicting AML prognosis based on VEN resistance-related senescence genes (VRSGs). In this study, 51 senescence genes were identified with VEN-resistance in AML. Using LASSO algorithms and multiple AML cohorts, a VEN-resistance senescence prognostic model (VRSP-M) was developed and validated based on 6-senescence genes. Results According to the median score of the signature, AMLs were classified into two subtypes. A worse prognosis and more adverse features occurred in the high-risk subtype, including older patients, non-de novo AML, poor cytogenetics, adverse risk of European LeukemiaNet (ELN) 2017 recommendation, and TP53 mutation. Patients in the high-risk subtype were mainly involved in monocyte differentiation, senescence, NADPH oxidases, and PD1 signaling pathway. The model's risk score was significantly associated with VEN-resistance, immune features, and immunotherapy response in AML. In vitro, the IC50 values of ABT-199 (VEN) rose progressively with increasing expression of G6PD and BAG3 in AML cell lines. Conclusions The 6-senescence genes prognostic model has significant meaning for the prediction of VEN-resistance, guiding personalized molecularly targeted therapies, and improving AML prognosis.
Collapse
Affiliation(s)
- Peng Ke
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Jundan Xie
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ting Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Meiyu Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yusha Guo
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Ying Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Huiying Qiu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zhao Zeng
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Suning Chen
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiebing Bao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
8
|
Mukohda M, Mizuno R, Ozaki H. Emerging evidence for a cardiovascular protective effect of concentrated Japanese plum juice. Hypertens Res 2023; 46:2428-2429. [PMID: 37532955 DOI: 10.1038/s41440-023-01395-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/04/2023] [Accepted: 07/08/2023] [Indexed: 08/04/2023]
Affiliation(s)
- Masashi Mukohda
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan.
| | - Risuke Mizuno
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| | - Hiroshi Ozaki
- Laboratory of Veterinary Pharmacology, Faculty of Veterinary Medicine, Okayama University of Science, Imabari, Japan
| |
Collapse
|
9
|
Cignarella A, Boscaro C, Albiero M, Bolego C, Barton M. Post-Transcriptional and Epigenetic Regulation of Estrogen Signaling. J Pharmacol Exp Ther 2023; 386:288-297. [PMID: 37391222 DOI: 10.1124/jpet.123.001613] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/17/2023] [Accepted: 06/16/2023] [Indexed: 07/02/2023] Open
Abstract
Post-translational and epigenetic regulation are important mechanisms controlling functions of genes and proteins. Although the "classic" estrogen receptors (ERs) have been acknowledged to function in mediating estrogen effects via transcriptional mechanisms, estrogenic agents modulate the turnover of several proteins via post-transcriptional and post-translational pathways including epigenetics. For instance, the metabolic and angiogenic action of G-protein coupled estrogen receptor (GPER) in vascular endothelial cells has been recently elucidated. By interacting with GPER, 17β-estradiol and the GPER agonist G1 enhance endothelial stability of 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3) and capillary tube formation by increasing ubiquitin-specific peptidase 19 levels, thereby reducing PFKFB3 ubiquitination and proteasomal degradation. In addition to ligands, the functional expression and trafficking of ERs can be modulated by post-translational modification, including palmitoylation. MicroRNAs (miRNAs), the most abundant form of endogenous small RNAs in humans, regulate multiple target genes and are at the center of the multi-target regulatory network. This review also discusses the emerging evidence of how miRNAs affect glycolytic metabolism in cancer, as well as their regulation by estrogens. Restoring dysregulated miRNA expression represents a promising strategy to counteract the progression of cancer and other disease conditions. Accordingly, estrogen post-transcriptional regulatory and epigenetic mechanisms represent novel targets for pharmacological and nonpharmacological intervention for the treatment and prevention of hormone-sensitive noncommunicable diseases, including estrogen-sensitive cancers of the reproductive system in women. SIGNIFICANCE STATEMENT: The effects of estrogen are mediated by several mechanisms that are not limited to the transcriptional regulation of target genes. Slowing down the turnover of master regulators of metabolism by estrogens allows cells to rapidly adapt to environmental cues. Identification of estrogen-targeted microRNAs may lead to the development of novel RNA therapeutics that disrupt pathological angiogenesis in estrogen-dependent cancers.
Collapse
Affiliation(s)
- Andrea Cignarella
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Carlotta Boscaro
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Mattia Albiero
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Chiara Bolego
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| | - Matthias Barton
- Departments of Medicine (A.C., Ca.B., M.A.) and Pharmaceutical and Pharmacological Sciences (Ch.B.), University of Padova, Padova, Italy; and Molecular Internal Medicine, University of Zürich and Andreas Grüntzig Foundation, Zürich, Switzerland (M.B.)
| |
Collapse
|
10
|
Kim SG, Sung JY, Kang YJ, Choi HC. Fisetin alleviates cellular senescence through PTEN mediated inhibition of PKCδ-NOX1 pathway in vascular smooth muscle cells. Arch Gerontol Geriatr 2023; 108:104927. [PMID: 36645971 DOI: 10.1016/j.archger.2023.104927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/29/2022] [Accepted: 01/08/2023] [Indexed: 01/13/2023]
Abstract
Reactive oxygen species (ROS) are a key risk factor of cellular senescence and age-related diseases, and protein kinase C (PKC) has been shown to activate NADPH oxidases (NOXs), which generate ROS. Although PKC activation induces oxidative stress, leading to the cellular dysfunction in various cell types, the correlation between PKC and senescence has not been reported in vascular smooth muscle cell (VSMC). Several studies have indicated cellular senescence is accompanied by phosphatase and tensin homolog (PTEN) loss and that an interaction exists between PTEN and PKC. Therefore, we aimed to determine whether PTEN and PKC are associated with VSMC senescence and to investigate the mechanism involved. We found hydrogen peroxide (H2O2) decreased PTEN expression and increased PKCδ phosphorylation. Moreover, H2O2 upregulated the NOX1 subunits, p22phox and p47phox, and induced VSMC senescence via p53-p21 signaling pathway. We identified PKCδ activation contributed to VSMC senescence through activation of NOX1 and ROS production. However, fisetin inhibited cellular senescence induced by the PTEN-PKCδ-NOX1-ROS signaling pathway, and this anti-aging effect was attributed to reduced ROS production caused by suppressing NOX1 activation. These results suggest that the PTEN-PCKδ signaling pathway is directly related to senescence via NOX1 activation and that the downregulation of PKCδ by flavonoids provides a potential means of treating age-associated diseases.
Collapse
Affiliation(s)
- Seul Gi Kim
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea; Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Jin Young Sung
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea; Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Young Jin Kang
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea
| | - Hyoung Chul Choi
- Department of Pharmacology, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea; Senotherapy-based Metabolic Disease Control Research Center, College of Medicine, Yeungnam University, 170 Hyunchung-Ro, Nam-Gu, Daegu 42415, Republic of Korea.
| |
Collapse
|
11
|
López-Acosta O, Ruiz-Ramírez A, Barrios-Maya MÁ, Alarcon-Aguilar J, Alarcon-Enos J, Céspedes Acuña CL, El-Hafidi M. Lipotoxicity, glucotoxicity and some strategies to protect vascular smooth muscle cell against proliferative phenotype in metabolic syndrome. Food Chem Toxicol 2023; 172:113546. [PMID: 36513245 DOI: 10.1016/j.fct.2022.113546] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 11/16/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022]
Abstract
Metabolic syndrome (MetS) is a risk factor for the development of cardiovascular disease (CVD) and atherosclerosis through a mechanism that involves vascular smooth muscle cell (VSMC) proliferation, lipotoxicity and glucotoxicity. Several molecules found to be increased in MetS, including free fatty acids, fatty acid binding protein 4, leptin, resistin, oxidized lipoprotein particles, and advanced glycation end products, influence VSMC proliferation. Most of these molecules act through their receptors on VSMCs by activating several signaling pathways associated with ROS generation in various cellular compartments. ROS from NADPH-oxidase and mitochondria have been found to promote VSMC proliferation and cell cycle progression. In addition, most of the natural or synthetic substances described in this review, including pharmaceuticals with hypoglycemic and hypolipidemic properties, attenuate VSMC proliferation by their simultaneous modulation of cell signaling and their scavenging property due to the presence of a phenolic ring in their structure. This review discusses recent data in the literature on the role that several MetS-related molecules and ROS play in the change from contractile to proliferative phenotype of VSMCs. Hence the importance of proposing an appropriate strategy to prevent uncontrolled VSMC proliferation using antioxidants, hypoglycemic and hypolipidemic agents.
Collapse
Affiliation(s)
- Ocarol López-Acosta
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Angélica Ruiz-Ramírez
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Miguel-Ángel Barrios-Maya
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico
| | - Javier Alarcon-Aguilar
- Laboratorio de Farmacología, Depto. de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana Unidad Iztapalapa, Iztapalapa, Mexico
| | - Julio Alarcon-Enos
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile
| | - Carlos L Céspedes Acuña
- Departamento de Ciencias Básicas, Facultad de Ciencias, Universidad del Bio Bio, Av. Andres Bello 720, Chillan, Chile.
| | - Mohammed El-Hafidi
- Depto de Biomedicina Cardiovascular, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano No 1, Colonia Sección XVI, Tlalpan, 14080, México D.F., Mexico.
| |
Collapse
|
12
|
Mitochondrial Dysfunction and Increased DNA Damage in Vascular Smooth Muscle Cells of Abdominal Aortic Aneurysm (AAA-SMC). OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6237960. [PMID: 36743698 PMCID: PMC9891816 DOI: 10.1155/2023/6237960] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/30/2022] [Accepted: 11/24/2022] [Indexed: 01/27/2023]
Abstract
There is increasing evidence for enhanced oxidative stress in the vascular wall of abdominal aortic aneurysms (AAA). Mitochondrial damage and dysfunction are hypothesized to be actors in altered production of reactive oxygen species (ROS) and oxidative stress. However, the role of mitochondria and oxidative stress in vascular remodelling and progression of AAA remains uncertain. We here addressed whether mitochondrial dysfunction is persistently increased in vascular smooth muscle cells (VSMCs) isolated from AAA compared to healthy VSMC. AAA-derived VSMC cultures (AAA-SMC, n = 10) and normal VSMC cultures derived from healthy donors (n = 7) were grown in vitro and analysed for four parameters, indicating mitochondrial dysfunction: (i) mitochondrial content and morphology, (ii) ROS production and antioxidative response, (iii) NADP+/NADPH content and ratio, and (iv) DNA damage, in the presence or absence of angiotensin II (AngII). AAA-SMC displayed increased mitochondrial circularity (rounded shape), reduced mitochondrial area, and reduced perimeter, indicating increased fragmentation and dysfunction compared to healthy controls. This was accompanied by significantly increased O2 - production, reduced NADP+/NADPH levels, a lower antioxidative response (indicated by antioxidative response element- (ARE-) driven luciferase reporter assays), more DNA damage (determined by percentage of γ-H2A.X-positive nuclei), and earlier growth arrest in AAA-SMC. Our data suggest that mitochondrial dysfunction and oxidative stress are persistently increased in AAA-SMC, emphasizing their implication in the pathophysiology of AAA.
Collapse
|
13
|
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
|
14
|
Sung JY, Kim SG, Kang YJ, Choi HC. Metformin mitigates stress-induced premature senescence by upregulating AMPKα at Ser485 phosphorylation induced SIRT3 expression and inactivating mitochondrial oxidants. Mech Ageing Dev 2022; 206:111708. [PMID: 35863470 DOI: 10.1016/j.mad.2022.111708] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 07/11/2022] [Accepted: 07/15/2022] [Indexed: 01/10/2023]
Abstract
The senescence of vascular smooth muscle cells (VSMCs) is an important cause of cardiovascular disease such as atherosclerosis and hypertension. These senescence may be triggered by many factors, such as oxidative stress, inflammation, DNA damage, and senescence-associated secretory phenotypes (SASPs). Mitochondrial oxidative stress induces cellular senescence, but the mechanisms by which mitochondrial reactive oxygen species (mtROS) regulates cellular senescence are still largely unknown. Here, we investigated the mechanism responsible for the anti-aging effect of metformin by examining links between VSMC senescence and mtROS in in vitro and in vivo. Metformin was found to increase p-AMPK (Ser485), but to decrease senescence-associated phenotypes and protein levels of senescence markers during ADR-induced VSMC senescence. Importantly, metformin decreased mtROS by inducing the deacetylation of superoxide dismutase 2 (SOD2) by increasing SIRT3 expression. Moreover, AMPK depletion reduced the expression of SIRT3 and increased the expression of acetylated SOD2 despite metformin treatment, suggesting AMPK activation by metformin is required to protect against mitochondrial oxidative stress by SIRT3. This study provides mechanistic evidence that metformin acts as an anti-aging agent and alleviates VSMC senescence by upregulating mitochondrial antioxidant induced p-AMPK (Ser485)-dependent SIRT3 expression, which suggests metformin has therapeutic potential for the treatment of age-associated vascular disease.
Collapse
Affiliation(s)
- Jin Young Sung
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Seul Gi Kim
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Young Jin Kang
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Hyoung Chul Choi
- Department of Pharmacology, College of Medicine, Yeungnam University, Daegu, Republic of Korea; Smart-aging Convergence Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea.
| |
Collapse
|
15
|
Quarleri J, Delpino MV. SARS-CoV-2 interacts with renin-angiotensin system: impact on the central nervous system in elderly patients. GeroScience 2022; 44:547-565. [PMID: 35157210 PMCID: PMC8853071 DOI: 10.1007/s11357-022-00528-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 02/08/2022] [Indexed: 01/18/2023] Open
Abstract
SARS-CoV-2 is a recently identified coronavirus that causes the current pandemic disease known as COVID-19. SARS-CoV-2 uses angiotensin-converting enzyme 2 (ACE2) as a receptor, suggesting that the initial steps of SARS-CoV-2 infection may have an impact on the renin-angiotensin system (RAS). Several processes are influenced by RAS in the brain. The neurological symptoms observed in COVID-19 patients, including reduced olfaction, meningitis, ischemic stroke, cerebral thrombosis, and delirium, could be associated with RAS imbalance. In this review, we focus on the potential role of disturbances in the RAS as a cause for central nervous system sequelae of SARS-CoV-2 infection in elderly patients.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus Y Sida (INBIRS), Universidad de Buenos Aires-CONICET, Paraguay 2155-Piso 11 (1121), Buenos Aires, Argentina.
| | - M Victoria Delpino
- Instituto de Investigaciones Biomédicas en Retrovirus Y Sida (INBIRS), Universidad de Buenos Aires-CONICET, Paraguay 2155-Piso 11 (1121), Buenos Aires, Argentina.
| |
Collapse
|
16
|
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: 85] [Impact Index Per Article: 28.3] [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
|
17
|
Herrmann J, Xia M, Gummi MR, Greco A, Schacke A, van der Giet M, Tölle M, Schuchardt M. Stressor-Induced "Inflammaging" of Vascular Smooth Muscle Cells via Nlrp3-Mediated Pro-inflammatory Auto-Loop. Front Cardiovasc Med 2022; 8:752305. [PMID: 34988126 PMCID: PMC8720922 DOI: 10.3389/fcvm.2021.752305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/29/2021] [Indexed: 01/14/2023] Open
Abstract
Calcification of the vessel wall as one structural pathology of aged vessels is associated with high cardiovascular mortality of elderly patients. Aging is linked to chronic sterile inflammation and high burden of reactive oxygen species (ROS), leading to activation of pattern recognition receptors (PRRs) such as Nlrp3 in vascular cells. The current study investigates the role of PRR activation in the calcification of vascular smooth muscle cells (VSMCs). Therefore, in vitro cell culture of primary rat VSMCs and ex vivo aortic stimulations were used to analyze osteogenic, senescence and inflammatory markers via real-time PCR, in situ RNA hybridization, Western Blot, photometric assays and histological staining. Induction of ROS and DNA-damage by doxorubicin induces a shift of VSMC phenotype toward the expression of osteogenic, senescence and inflammatory proteins. Induction of calcification is dependent on Nlrp3 activity. Il-1β as a downstream target of Nlrp3 induces the synthetic, pro-calcifying VSMC phenotype. Inhibition of PRR with subsequent reduction of chronic inflammation might be an interesting target for reduction of calcification of VSMCs, with subsequent reduction of cardiovascular mortality of patients suffering from vessel stiffness.
Collapse
Affiliation(s)
- Jaqueline Herrmann
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany.,Department of Chemistry, Biochemistry and Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Mengdi Xia
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany.,Department of Nephrology, Second Clinical Medical Institution of North Sichuan Medical College (Nanchong Central Hospital), Nanchong, China
| | - Manasa Reddy Gummi
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Anna Greco
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Annika Schacke
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Markus van der Giet
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Markus Tölle
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| | - Mirjam Schuchardt
- Department of Nephrology and Medical Intensive Care, Charité - Universitätsmedizin Berlin, Cooperate Member of Freie Universität and Humboldt Universität, Berlin, Germany
| |
Collapse
|
18
|
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: 2.0] [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
|
19
|
Hung YC, Liu YC, Wu BN, Yeh JL, Hsu JH. Cinaciguat Prevents Postnatal Closure of Ductus Arteriosus by Vasodilation and Anti-remodeling in Neonatal Rats. Front Physiol 2021; 12:661171. [PMID: 34393808 PMCID: PMC8358454 DOI: 10.3389/fphys.2021.661171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 07/06/2021] [Indexed: 12/19/2022] Open
Abstract
Closure of the ductus arteriosus (DA) involves vasoconstriction and vascular remodeling. Cinaciguat, a soluble guanylyl cyclase (sGC) activator, was reported with vasodilatory and anti-remodeling effects on pulmonary hypertensive vessels. However, its effects on DA are not understood. Therefore, we investigated whether cinaciguat regulated DA patency and examined its underlying mechanisms. In vivo, we found that cinaciguat (10 mg/kg, i.p. at birth) prevented DA closure at 2 h after birth with luminal patency and attenuated intimal thickening. These anti-remodeling effects were associated with enhanced expression of cyclic guanosine monophosphate (cGMP) in DA. Ex vivo, cinaciguat dilated oxygen-induced DA constriction dose-dependently. Such vasodilatory effect was blunted by KT-5823, a PKG inhibitor. In DA smooth muscle cells (DASMCs), we further showed that cinaciguat inhibited angiotensin II (Ang II)-induced proliferation and migration of DASMCs. In addition, cinaciguat inhibited Ang II-induced mitochondrial reactive oxygen species (ROS) production. Finally, Ang II-activated MAPKs and Akt were also inhibited by cinaciguat. In conclusion, cinaciguat prevents postnatal DA closure by vasodilation and anti-remodeling through the cGMP/PKG pathway. The mechanisms underlying anti-remodeling effects include anti-proliferation and anti-migration, with attenuation of mitochondrial ROS production, MAPKs, and Akt signaling. Thus, this study implicates that sGC activation may be a promising novel strategy to regulate DA patency.
Collapse
Affiliation(s)
- Yu-Chi Hung
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pediatrics, St. Joseph Hospital, Kaohsiung, Taiwan
| | - Yi-Ching Liu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Bin-Nan Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jwu-Lai Yeh
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pharmacology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Medical Research, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Jong-Hau Hsu
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
20
|
Abstract
Cells respond to stress by activating a variety of defense signaling pathways, including cell survival and cell death pathways. Although cell survival signaling helps the cell to recover from acute insults, cell death or senescence pathways induced by chronic insults can lead to unresolved pathologies. Arterial hypertension results from chronic physiological maladaptation against various stressors represented by abnormal circulating or local neurohormonal factors, mechanical stress, intracellular accumulation of toxic molecules, and dysfunctional organelles. Hypertension and aging share common mechanisms that mediate or prolong chronic cell stress, such as endoplasmic reticulum stress and accumulation of protein aggregates, oxidative stress, metabolic mitochondrial stress, DNA damage, stress-induced senescence, and proinflammatory processes. This review discusses common adaptive signaling mechanisms against these stresses including unfolded protein responses, antioxidant response element signaling, autophagy, mitophagy, and mitochondrial fission/fusion, STING (signaling effector stimulator of interferon genes)-mediated responses, and activation of pattern recognition receptors. The main molecular mechanisms by which the vasculature copes with hypertensive and aging stressors are presented and recent advancements in stress-adaptive signaling mechanisms as well as potential therapeutic targets are discussed.
Collapse
Affiliation(s)
- Stephanie M. Cicalese
- These authors contributed equally and are considered co-first authors
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Josiane Fernandes da Silva
- These authors contributed equally and are considered co-first authors
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| | - Fernanda Priviero
- These authors contributed equally and are considered co-first authors
- Cardiovascular Translational Research Center and Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina, USA
| | - R. Clinton Webb
- Cardiovascular Translational Research Center and Department of Cell Biology and Anatomy, University of South Carolina, Columbia, South Carolina, USA
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania, USA
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil
| |
Collapse
|
21
|
Liu D, Sun WP, Chen JW, Jiang Y, Xue R, Wang LH, Murao K, Zhang GX. Autophagy contributes to angiotensin II induced dysfunction of HUVECs. Clin Exp Hypertens 2021; 43:462-473. [PMID: 33775188 DOI: 10.1080/10641963.2021.1901110] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
BACKGROUND Signal transduction of Angiotensin II (Ang II) induced autophagy and its role in Ang II-induced dysfunction of HUVECs are still unclear. METHODS HUVECs are stimulated with different doses of Ang II (10-9-10-5 mol/L) for different time (6-48 hours). Autophagy-related protein markers: LC3, Beclin-1 and SQSTM1/p62 are measured by western blot. RESULTS Incubation with Ang II increases autophagic flux (Beclin-1, autophagosomes formation, and degradation of SQSTM1/p62, LC3-I). Increased autophagic levels are inhibited by pretreatment with Ang II type 1 receptor (AT1) blocker (Candesartan), NADPH Oxidase inhibitor (apocycin), mitochondrial KATP channels inhibitor (5-hydroxydecanoate, 5HD). 3-Methyladenine (inhibitors of autophagy) and rapamycin (activator of autophagy) respectively inhibits or activates Ang II-induced autophagy levels. Ang II decreases phosphorylation of endothelial nitric oxide synthase (eNOS) and NO production in HUVECs. L-NAME (NOS inhibitor) totally mimics the actions of Ang II on eNOS, NO production and autophagy levels. Rapamycin further decreases NO production combined with Ang II. Silence Atg5 completely reverses Ang II-activated autophagy levels. CONCLUSIONS Our results demonstrate that Ang II stimulation increases autophagy levels via AT1 receptor, NADPH oxidase, mitochondrial KATP channel, eNOS, Atg5 signal pathway in HUVECs, and activation of autophagy contributes to Ang II induced dysfunction of HUVECs.
Collapse
Affiliation(s)
- Di Liu
- Department of Physiology and Neuroscience, Soochow University - Dushu Lake Campus, Suzhou, China
| | - Wan-Pin Sun
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Pharmaceutical Science, Laboratory of Molecular Diagnostics, Medical College of Soochow University, Suzhou, P.R. China
| | - Jing-Wei Chen
- Department of Internal Medicine, the Affiliated Suzhou Chinese Traditional Medicine Hospital, Nanjing University of Chinese Medicine, Suzhou, P.R. China
| | - Yan Jiang
- Department of Physiology and Neuroscience, Soochow University - Dushu Lake Campus, Suzhou, China
| | - Rong Xue
- Department of Physiology and Neuroscience, Soochow University - Dushu Lake Campus, Suzhou, China
| | - Lin-Hui Wang
- Department of Physiology and Neuroscience, Soochow University - Dushu Lake Campus, Suzhou, China
| | - Koji Murao
- Department of Clinical Laboratory, Faculty of Medicine, Kagawa University, Kagawa, Japan
| | - Guo-Xing Zhang
- Department of Physiology and Neuroscience, Soochow University - Dushu Lake Campus, Suzhou, China
| |
Collapse
|
22
|
Cicalese S, Okuno K, Eguchi S. Detection of Protein Aggregation and Proteotoxicity Induced by Angiotensin II in Vascular Smooth Muscle Cells. J Cardiovasc Pharmacol 2021; 77:43-48. [PMID: 33079831 DOI: 10.1097/fjc.0000000000000934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 10/06/2020] [Indexed: 12/26/2022]
Abstract
ABSTRACT Disruption of protein quality control occurs with aging and cardiovascular pathologies including arterial stiffness and hypertension. Angiotensin II (Ang II) is believed to induce endoplasmic reticulum stress in vascular smooth muscle cells (VSMCs), thus contributing to vascular remodeling and dysfunction. However, whether Ang II increases formation of protein aggregates and mediates proteotoxicity in VSMCs remain obscure. Accordingly, this study aimed to establish a quantitative method of protein aggregate detection induced by Ang II and to investigate their potential involvement in inflammatory and senescence responses. Proteostat staining showed increased aggregate numbers per cell on Ang II exposure. Immunoblot analysis further showed an increase in preamyloid oligomer presence in a detergent insoluble protein fraction purified from VSMCs stimulated with Ang II. Moreover, these responses were attenuated by treatment with chemical chaperone, 4-phenylbutyrate. 4-phenylbutyrate further blocked Ang II-induced senescence associated β-galactosidase activity and THP-1 monocyte adhesion in VSMCs. These data suggest that Ang II induces proteotoxicity in VSMCs which likely contributes to aging and inflammation in the vasculature.
Collapse
MESH Headings
- Angiotensin II/toxicity
- Animals
- Cell Adhesion/drug effects
- Cellular Senescence/drug effects
- Endoplasmic Reticulum Stress/drug effects
- Humans
- Male
- Monocytes/drug effects
- Monocytes/metabolism
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/drug effects
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Protein Aggregates
- Protein Aggregation, Pathological
- Rats, Sprague-Dawley
- THP-1 Cells
- Rats
Collapse
Affiliation(s)
- Stephanie Cicalese
- Department of Physiology, Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA
| | | | | |
Collapse
|
23
|
Cosarderelioglu C, Nidadavolu LS, George CJ, Oh ES, Bennett DA, Walston JD, Abadir PM. Brain Renin-Angiotensin System at the Intersect of Physical and Cognitive Frailty. Front Neurosci 2020; 14:586314. [PMID: 33117127 PMCID: PMC7561440 DOI: 10.3389/fnins.2020.586314] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 08/25/2020] [Indexed: 12/15/2022] Open
Abstract
The renin–angiotensin system (RAS) was initially considered to be part of the endocrine system regulating water and electrolyte balance, systemic vascular resistance, blood pressure, and cardiovascular homeostasis. It was later discovered that intracrine and local forms of RAS exist in the brain apart from the endocrine RAS. This brain-specific RAS plays essential roles in brain homeostasis by acting mainly through four angiotensin receptor subtypes; AT1R, AT2R, MasR, and AT4R. These receptors have opposing effects; AT1R promotes vasoconstriction, proliferation, inflammation, and oxidative stress while AT2R and MasR counteract the effects of AT1R. AT4R is critical for dopamine and acetylcholine release and mediates learning and memory consolidation. Consequently, aging-associated dysregulation of the angiotensin receptor subtypes may lead to adverse clinical outcomes such as Alzheimer’s disease and frailty via excessive oxidative stress, neuroinflammation, endothelial dysfunction, microglial polarization, and alterations in neurotransmitter secretion. In this article, we review the brain RAS from this standpoint. After discussing the functions of individual brain RAS components and their intracellular and intracranial locations, we focus on the relationships among brain RAS, aging, frailty, and specific neurodegenerative diseases, such as Alzheimer’s disease, Parkinson’s disease, and vascular cognitive impairment, through oxidative stress, neuroinflammation, and vascular dysfunction. Finally, we discuss the effects of RAS-modulating drugs on the brain RAS and their use in novel treatment approaches.
Collapse
Affiliation(s)
- Caglar Cosarderelioglu
- Division of Geriatrics, Department of Internal Medicine, Ankara University School of Medicine, Ankara, Turkey.,Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Lolita S Nidadavolu
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Claudene J George
- Division of Geriatrics, Department of Medicine, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, NY, United States
| | - Esther S Oh
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL, United States
| | - Jeremy D Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter M Abadir
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
24
|
Okuno K, Cicalese S, Elliott KJ, Kawai T, Hashimoto T, Eguchi S. Targeting Molecular Mechanism of Vascular Smooth Muscle Senescence Induced by Angiotensin II, A Potential Therapy via Senolytics and Senomorphics. Int J Mol Sci 2020; 21:ijms21186579. [PMID: 32916794 PMCID: PMC7555774 DOI: 10.3390/ijms21186579] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is a prevalent issue in the global aging population. Premature vascular aging such as elevated arterial stiffness appears to be a major risk factor for CVD. Vascular smooth muscle cells (VSMCs) are one of the essential parts of arterial pathology and prone to stress-induced senescence. The pervasiveness of senescent VSMCs in the vasculature increases with age and can be further expedited by various stressing events such as oxidative stress, mitochondria dysfunction, endoplasmic reticulum stress, and chronic inflammation. Angiotensin II (AngII) can induce many of these responses in VSMCs and is thus considered a key regulator of VSMC senescence associated with CVD. Understanding the precise mechanisms and consequences of senescent cell accumulation may uncover a new generation of therapies including senolytic and senomorphic compounds against CVD. Accordingly, in this review article, we discuss potential molecular mechanisms of VSMC senescence such as those induced by AngII and the therapeutic manipulations of senescence to control age-related CVD and associated conditions such as by senolytic.
Collapse
Affiliation(s)
- Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Stephanie Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Katherine J. Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Tomoki Hashimoto
- Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA;
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
- Correspondence:
| |
Collapse
|
25
|
Cross-Talk between NADPH Oxidase and Mitochondria: Role in ROS Signaling and Angiogenesis. Cells 2020; 9:cells9081849. [PMID: 32781794 PMCID: PMC7466096 DOI: 10.3390/cells9081849] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/27/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis, a new vessel formation from the pre-existing ones, is essential for embryonic development, wound repair and treatment of ischemic heart and limb diseases. However, dysregulated angiogenesis contributes to various pathologies such as diabetic retinopathy, atherosclerosis and cancer. Reactive oxygen species (ROS) derived from NADPH oxidase (NOX) as well as mitochondria play an important role in promoting the angiogenic switch from quiescent endothelial cells (ECs). However, how highly diffusible ROS produced from different sources and location can communicate with each other to regulate angiogenesis remains unclear. To detect a localized ROS signal in distinct subcellular compartments in real time in situ, compartment-specific genetically encoded redox-sensitive fluorescence biosensors have been developed. Recently, the intercellular communication, “cross-talk”, between ROS derived from NOX and mitochondria, termed “ROS-induced ROS release”, has been proposed as a mechanism for ROS amplification at distinct subcellular compartments, which are essential for activation of redox signaling. This “ROS-induced ROS release” may represent a feed-forward mechanism of localized ROS production to maintain sustained signaling, which can be targeted under pathological conditions with oxidative stress or enhanced to promote therapeutic angiogenesis. In this review, we summarize the recent knowledge regarding the role of the cross-talk between NOX and mitochondria organizing the sustained ROS signaling involved in VEGF signaling, neovascularization and tissue repair.
Collapse
|
26
|
Mogi M. Effect of renin-angiotensin system on senescence. Geriatr Gerontol Int 2020; 20:520-525. [PMID: 32346971 DOI: 10.1111/ggi.13927] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/24/2020] [Accepted: 03/04/2020] [Indexed: 12/11/2022]
Abstract
The renin-angiotensin system (RAS) plays crucial roles in the control of blood pressure and sodium homeostasis. Moreover, RAS also acts as a key player in cell and organ senescence, mainly by activation of the classical axis of angiotensin (Ang) converting enzyme (ACE)/Ang II/Ang II type 1 receptor via overproduction of reactive oxygen species. Overactivation of the classical RAS axis induces organ dysfunction in the vasculature, brain, kidney and skeletal muscle, resulting in atherosclerosis, stroke, chronic kidney disease and sarcopenia. Moreover, RAS has been shown to regulate lifespan, using gene-modification models. Recently, mice lacking the Ang II type 1 receptor were shown to exhibit an increase in lifespan compared with control mice. Here, the effect of RAS on age-related tissue dysfunction in several organs is reviewed, including not only the classical axis but also protective functions of RAS such as the ACE2/Ang (1-7)/Mas axis. Geriatr Gerontol Int 2020; ••: ••-••.
Collapse
Affiliation(s)
- Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Ehime, Japan
| |
Collapse
|
27
|
AT1R-Mediated Apoptosis of Bone Marrow Mesenchymal Stem Cells Is Associated with mtROS Production and mtDNA Reduction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4608165. [PMID: 31772704 PMCID: PMC6854225 DOI: 10.1155/2019/4608165] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 04/18/2019] [Accepted: 04/23/2019] [Indexed: 01/02/2023]
Abstract
Angiotensin II (Ang II) is used as an inducer for the differentiation of mesenchymal stem cells (MSCs). Whether the commonly used doses of Ang II for MSC differentiation affect cell apoptosis has not been elucidated. In this study, we investigated the effect of Ang II on the apoptosis of bone marrow MSCs (BMMSCs), and its relations to the activation of Ang II receptor-1- (AT1R-) signaling, mitochondrial ROS (mtROS) generation, and mitochondrial DNA (mtDNA) leakage. AT1R expression in BMMSCs was identified by immunostaining and Western-blotting assays. BMMSC viability was measured by MTT assay following exposure to 1 nM~1 mM Ang II for 12 hours. Cell apoptosis, mtROS, and mtDNA levels were detected by FAM-FLICA® Poly Caspase, MitoSOX™ superoxide, and PicoGreen staining, respectively. The expressions of Bcl2 and Bax were measured by Western-blotting assays. Next, we used losartan to block AT1R-signaling and subsequently measured apoptosis, mtROS, and mtDNA levels, again. The maximum viability of BMMSCs was in response to 100 nM Ang II, after that it began to decrease with the increase of Ang II doses, indicating that Ang II (≧1 μM) may cause apoptosis of BMMSCs. As expected, 1 μM and 10 μM Ang II both caused BMMSC apoptosis. Furthermore, 1 μM and 10 μM Ang II could also induce mtROS generation and cause a marked mtDNA leakage. The application of losartan markedly inhibited Ang II-induced mtROS production, mtDNA leakage, and BMMSC apoptosis. In conclusion, the activation of AT1R-signaling stimulates apoptosis of BMMSCs, which is associated mtROS production and mtDNA reduction.
Collapse
|
28
|
Kim I, Park CS, Lee HY. Angiotensin II Type 1 Receptor Blocker, Fimasartan, Reduces Vascular Smooth Muscle Cell Senescence by Inhibiting the CYR61 Signaling Pathway. Korean Circ J 2019; 49:615-626. [PMID: 31074217 PMCID: PMC6597448 DOI: 10.4070/kcj.2018.0379] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Revised: 12/26/2018] [Accepted: 01/23/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Angiotensin II (Ang II) has been suggested to accelerate vascular senescence, however the molecular mechanism(s) remain unknown. METHODS We cultured human coronary artery smooth muscle cells (hCSMCs) and treated Ang II and/or fimasartan. Or we transfected adenoviral vectors expressing CYR61 (Ad-CYR61) or antisense CYR61 (Ad-As-CYR61). Cellular senescence was evaluated senescence-associated β-galactosidase (SA-β-gal) assay. The molecular mechanisms were investigated real-time PCR and western blots. RESULTS SA-β-gal-positive cells significantly increased in Ang II-treated hCSMCs (5.77±1.43-fold compared with the control). The effect of Ang II was significantly attenuated by pretreatment with the Ang II type 1 receptor blocker, fimasartan (2.00±0.92-fold). The expression of both p53 and p16 senescence regulators was significantly increased by Ang II (p53: 1.39±0.17, p16: 1.19±0.10-fold vs. the control), and inhibited by fimasartan. Cysteine-rich angiogenic protein 61 (CYR61) was rapidly induced by Ang II. Compared with the control, Ad-CYR61-transfected hCSMCs showed significantly increased SA-β-gal-positive cells (3.47±0.65-fold). Upon transfecting Ad-AS-CYR61, Ang II-induced senescence (3.74±0.23-fold) was significantly decreased (1.77±0.60-fold). p53 expression by Ang II was significantly attenuated by Ad-AS-CYR61, whereas p16 expression was not regulated. Ang II activated ERK1/2 and p38 MAPK, which was significantly blocked by fimasartan. ERK and p38 inhibition both regulated Ang II-induced CYR61 expression. However, p53 expression was only regulated by ERK1/2, whereas p16 expression was only attenuated by p38 MAPK. CONCLUSIONS Ang II induced vascular senescence by the ERK/p38 MAPK-CYR61 pathway and ARB, fimasartan, protected against Ang II-induced vascular senescence.
Collapse
Affiliation(s)
- Inho Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Microbiology and Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Chan Soon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Hae Young Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
29
|
Vascular smooth muscle cell senescence and age-related diseases: State of the art. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1810-1821. [PMID: 31109451 DOI: 10.1016/j.bbadis.2018.08.015] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 07/20/2018] [Accepted: 08/13/2018] [Indexed: 02/07/2023]
|
30
|
Choi H, Stark RJ, Raja BS, Dikalova A, Lamb FS. Apoptosis signal-regulating kinase 1 activation by Nox1-derived oxidants is required for TNFα receptor endocytosis. Am J Physiol Heart Circ Physiol 2019; 316:H1528-H1537. [PMID: 30925081 DOI: 10.1152/ajpheart.00741.2018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tumor necrosis factor-α (TNFα) is a proinflammatory cytokine that is closely linked to the development of cardiovascular disease. TNFα activates NADPH oxidase 1 (Nox1) and reactive oxygen species (ROS), including superoxide (O2·-), production extracellularly is required for subsequent signaling in vascular smooth muscle cells (VSMCs). Apoptosis signal-regulating kinase 1 (ASK1) is a mitogen-activated protein kinase kinase kinase that is activated by oxidation of associated thioredoxin. The role of ASK1 in Nox1-mediated signaling by TNFα is poorly defined. We hypothesized that ASK1 is required for TNFα receptor endocytosis and subsequent inflammatory TNFα signaling. We employed a knockdown strategy to explore the role of ASK1 in TNFα signaling in VSMCs. siRNA targeting ASK1 had no effect on TNFα-induced extracellular O2·- production. However, siASK1 inhibited receptor endocytosis as well as phosphorylation of two endocytosis-related proteins, dynamin1 and caveolin1. Intracellular O2·- production was subsequently reduced, as were other inflammatory signaling steps including NF-κB activation, IL-6 production, inducible nitric oxide synthase and VCAM expression, and VSMC proliferation. Prolonged exposure to TNFα (24 h) increased tumor necrosis factor receptor (TNFR) subtype 1 and 2 expression, and these effects were also attenuated by siASK1. ASK1 coimmunoprecipitated with both Nox1 and the leucine rich repeat containing 8A anion channel, two essential components of the TNFR1 signaling complex. Activation of ASK1 by autophosphorylation at Thr845 occurs following thioredoxin dissociation, and this requires the presence of Nox1. Thus, Nox1 is part of the multiprotein ASK1 signaling complex. In response to TNFα, ASK1 is activated by Nox1-derived oxidants, and this plays a critical role in translating these ROS into a physiologic response in VSMCs. NEW & NOTEWORTHY Apoptosis signal-regulating kinase 1 (ASK1) drives dynamin1 and caveolin1 phosphorylation and TNFα receptor endocytosis. ASK1 modulates TNFα-induced NF-κB activation, survival, and proliferation. ASK1 and NADPH oxidase 1 (Nox1) physically associate in a multiprotein signaling complex. Nox1 is required for TNFα-induced ASK1 activation.
Collapse
Affiliation(s)
- Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Ryan J Stark
- Department of Pediatrics, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Benjamin S Raja
- Department of Pediatrics, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Anna Dikalova
- Department of Pediatrics, Vanderbilt University Medical Center , Nashville, Tennessee
| | - Fred S Lamb
- Department of Pediatrics, Vanderbilt University Medical Center , Nashville, Tennessee
| |
Collapse
|
31
|
Li S, Zhan JK, Wang YJ, Lin X, Zhong JY, Wang Y, Tan P, He JY, Cui XJ, Chen YY, Huang W, Liu YS. Exosomes from hyperglycemia-stimulated vascular endothelial cells contain versican that regulate calcification/senescence in vascular smooth muscle cells. Cell Biosci 2019; 9:1. [PMID: 30622695 PMCID: PMC6317223 DOI: 10.1186/s13578-018-0263-x] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022] Open
Abstract
Background To determine whether and how exosomes from human umbilical vein endothelial cells (HUVEC-Exos) regulates vascular smooth muscle cells (VSMCs) calcification/senescence in high glucose condition. Methods HUVEC-Exos were isolated from normal glucose (NG) and high glucose (HG) stimulated HUVECs (NG/HG-HUVEC-Exos) by super speed centrifugation. HUVEC-Exos were identified by transmission electron microscopy and Western blot of CD63. Protein profile in HUVEC-Exos was examined to screen the candidate molecules that mediate HUVEC-Exos function. VSMCs were incubated with HUVEC-Exos. A series of functional assays in vitro were performed to assess the effects of HUVEC-Exos on the calcification/senescence of VSMCs. The role of the candidate protein in HUVEC-Exos-induced VSMCs dysfunction was assessed. Results Exosomes isolated from HG-HUVEC-Exos induced calcification/senescence in VSMCs as assessed by Alizarin Red Staining, senescence-associated β-galactosidase (SA-β-gal) staining, and the expression of ALP and p21. HG-HUVEC-Exos significantly increased LDH activity, as well as the product of lipid peroxidation (MDA content), and decreased oxidative stress marker activity, as compared with NG-HUVEC-Exos. Moreover, mechanism studies showed that mitochondrial membrane potential and the expression levels of mitochondrial function related protein HADHA and Cox-4 were significantly decreased in HG-HUVEC-Exos compared to controls. Proteomic analysis showed that HG-HUVEC-Exos consisted of higher level of versican (VCAN), as compared with NG-HUVEC-Exos. Observation under laser confocal microscopy revealed that most green fluorescence of VCAN could overlap with the red fluorescence came from mitochondria, indicating VCAN is mainly localized to the mitochondria of VSMCs. Knockdown of VCAN with siRNA in HUVECs, inhibited HG-HUVEC-Exos-induced mitochondrial dysfunction and calcification/senescence of VSMCs. Conclusions Our data indicate an intracellular role for VCAN in VSMCs. VCAN participates in hyperglycemia-induced calcification/senescence via modulation of mitochondrial function in VSMCs. Electronic supplementary material The online version of this article (10.1186/s13578-018-0263-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Shuang Li
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Jun-Kun Zhan
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Yan-Jiao Wang
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Xiao Lin
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Jia-Yu Zhong
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Yi Wang
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Pan Tan
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Jie-Yu He
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Xing-Jun Cui
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Yi-Yin Chen
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - Wu Huang
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| | - You-Shuo Liu
- Department of Geriatrics, Institute of Aging and Geriatrics, The Second Xiangya Hospital, Central South University, 139 Renmin Road, Changsha, 410011 Hunan People's Republic of China
| |
Collapse
|
32
|
Forrester SJ, Booz GW, Sigmund CD, Coffman TM, Kawai T, Rizzo V, Scalia R, Eguchi S. Angiotensin II Signal Transduction: An Update on Mechanisms of Physiology and Pathophysiology. Physiol Rev 2018; 98:1627-1738. [PMID: 29873596 DOI: 10.1152/physrev.00038.2017] [Citation(s) in RCA: 718] [Impact Index Per Article: 102.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin-aldosterone system plays crucial roles in cardiovascular physiology and pathophysiology. However, many of the signaling mechanisms have been unclear. The angiotensin II (ANG II) type 1 receptor (AT1R) is believed to mediate most functions of ANG II in the system. AT1R utilizes various signal transduction cascades causing hypertension, cardiovascular remodeling, and end organ damage. Moreover, functional cross-talk between AT1R signaling pathways and other signaling pathways have been recognized. Accumulating evidence reveals the complexity of ANG II signal transduction in pathophysiology of the vasculature, heart, kidney, and brain, as well as several pathophysiological features, including inflammation, metabolic dysfunction, and aging. In this review, we provide a comprehensive update of the ANG II receptor signaling events and their functional significances for potential translation into therapeutic strategies. AT1R remains central to the system in mediating physiological and pathophysiological functions of ANG II, and participation of specific signaling pathways becomes much clearer. There are still certain limitations and many controversies, and several noteworthy new concepts require further support. However, it is expected that rigorous translational research of the ANG II signaling pathways including those in large animals and humans will contribute to establishing effective new therapies against various diseases.
Collapse
Affiliation(s)
- Steven J Forrester
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - George W Booz
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Curt D Sigmund
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Thomas M Coffman
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Victor Rizzo
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Rosario Scalia
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University , Philadelphia, Pennsylvania ; Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center , Jackson, Mississippi ; Department of Pharmacology, Center for Hypertension Research, Roy J. and Lucille A. Carver College of Medicine, University of Iowa , Iowa City, Iowa ; and Duke-NUS, Singapore and Department of Medicine, Duke University Medical Center , Durham, North Carolina
| |
Collapse
|
33
|
Ji S, Zheng Z, Liu S, Ren G, Gao J, Zhang Y, Li G. Resveratrol promotes oxidative stress to drive DLC1 mediated cellular senescence in cancer cells. Exp Cell Res 2018; 370:292-302. [DOI: 10.1016/j.yexcr.2018.06.031] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 12/20/2022]
|
34
|
A causal link between oxidative stress and inflammation in cardiovascular and renal complications of diabetes. Clin Sci (Lond) 2018; 132:1811-1836. [PMID: 30166499 DOI: 10.1042/cs20171459] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/22/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Chronic renal and vascular oxidative stress in association with an enhanced inflammatory burden are determinant processes in the development and progression of diabetic complications including cardiovascular disease (CVD), atherosclerosis and diabetic kidney disease (DKD). Persistent hyperglycaemia in diabetes mellitus increases the production of reactive oxygen species (ROS) and activates mediators of inflammation as well as suppresses antioxidant defence mechanisms ultimately contributing to oxidative stress which leads to vascular and renal injury in diabetes. Furthermore, there is increasing evidence that ROS, inflammation and fibrosis promote each other and are part of a vicious connection leading to development and progression of CVD and kidney disease in diabetes.
Collapse
|
35
|
Activating the PGC-1 α/TERT Pathway by Catalpol Ameliorates Atherosclerosis via Modulating ROS Production, DNA Damage, and Telomere Function: Implications on Mitochondria and Telomere Link. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2876350. [PMID: 30046372 PMCID: PMC6036816 DOI: 10.1155/2018/2876350] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Accepted: 03/15/2018] [Indexed: 12/17/2022]
Abstract
Catalpol, an iridoid glucoside, has been found present in large quantities in the root of Rehmannia glutinosa L. and showed a strong antioxidant capacity in the previous study. In the present work, the protective effect of catalpol against AS via inhibiting oxidative stress, DNA damage, and telomere shortening was found in LDLr-/- mice. This study also shows that activation of the peroxisome proliferator-activated receptor-γ coactivator-1α (PGC-1α)/telomerase reverse transcriptase (TERT) pathway, which is the new link between mitochondria and telomere, was involved in the protective effects of catalpol. Further, by using PGC-1α or TERT siRNA in oxLDL-treated macrophages, it is proved that catalpol reduced oxidative stress, telomere function, and related DNA damage at least partly through activating the PGC-1α/TERT pathway. Moreover, dual luciferase activity assay-validated catalpol directly enhanced PGC-1α promoter activity. In conclusion, our study revealed that the PGC-1α/TERT pathway might be a possible therapeutic target in AS and catalpol has highly favorable characteristics for the treatment of AS via modulating this pathway.
Collapse
|
36
|
Zhao M, Bai M, Ding G, Zhang Y, Huang S, Jia Z, Zhang A. Angiotensin II Stimulates the NLRP3 Inflammasome to Induce Podocyte Injury and Mitochondrial Dysfunction. KIDNEY DISEASES 2018; 4:83-94. [PMID: 29998123 DOI: 10.1159/000488242] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 02/05/2018] [Indexed: 12/19/2022]
Abstract
Background We previously reported that the NLRP3 inflammasome played an important role in mediating the podocyte injury induced by aldosterone. However, more studies on the role of the NLRP3 inflammasome in the pathogenesis of podocytopathy are still required. The present study was undertaken to study the role of the NLRP3 inflammasome in angiotensin II (Ang II)-induced podocyte injury, as well as the potential mechanisms. Methods In this study, we used an Ang II infusion model in NLRP3-/- mice. In cultured podocytes, we used siRNA to silence NLRP3; then we treated the podocytes with Ang II. Results Following Ang II treatment, we found that the NLRP3 inflammasome was significantly activated in line with mitochondrial dysfunction in a dose- and time-dependent manner. Silencing NLRP3 by siRNA transfection ameliorated podocyte apoptosis, attenuated the loss of the podocyte proteins nephrin and podocin, and protected mitochondrial function. Ang II infusion activated the NLRP3 inflammasome, caused albuminuria, and induced podocyte damage, which was all blocked in the NLRP3-/- mice. At the same time, NLRP3 deletion also ameliorated the mitochondrial dysfunction induced by Ang II infusion. However, the deletion of NLRP3 did not affect the Ang II hypertension. Conclusion Taken together, these results demonstrate an important role of the NLRP3 inflammasome in mediating Ang II-induced podocyte injury and mitochondrial dysfunction, suggesting that the NLRP3 inflammasome might be an effective therapeutic target against podocytopathy.
Collapse
Affiliation(s)
- Min Zhao
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Mi Bai
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Guixia Ding
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Yue Zhang
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Songming Huang
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China
| | - Zhanjun Jia
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Nanjing, China
| | - Aihua Zhang
- Department of Nephrology, State Key Laboratory of Reproductive Medicine, Children's Hospital of Nanjing Medical University, Nanjing, China.,Jiangsu Key Laboratory of Pediatrics, Nanjing Medical University, Nanjing, China.,Nanjing Key Laboratory of Pediatrics, Nanjing, China
| |
Collapse
|
37
|
NADPH Oxidases and Mitochondria in Vascular Senescence. Int J Mol Sci 2018; 19:ijms19051327. [PMID: 29710840 PMCID: PMC5983750 DOI: 10.3390/ijms19051327] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/22/2018] [Accepted: 04/27/2018] [Indexed: 02/07/2023] Open
Abstract
Aging is the major risk factor in the development of cardiovascular diseases (CVDs), including hypertension, atherosclerosis, and myocardial infarction. Oxidative stress caused by overproduction of reactive oxygen species (ROS) and/or by reduced expression of antioxidant enzymes is a major contributor to the progression of vascular senescence, pathologic remodeling of the vascular wall, and disease. Both oxidative stress and inflammation promote the development of senescence, a process by which cells stop proliferating and become dysfunctional. This review focuses on the role of the mitochondria and the nicotinamide adenine dinucleotide phosphate (NADPH) oxidases Nox1 and Nox4 in vascular senescence, and their contribution to the development of atherosclerosis. Recent findings are reviewed, supporting a critical role of the mitochondrial regulator peroxisome proliferator-activated receptor gamma (PPARγ) coactivator-1α (PGC-1α), the inflammatory gene nuclear factor κB (NF-κB), zinc, the zinc transporters (ZnTs) ZnT3 and ZnT10, and angiotensin II (Ang II) in mitochondrial function, and their role in telomere stability, which provides new mechanistic insights into a previously proposed unified theory of aging.
Collapse
|
38
|
De Luca M. The role of the cell-matrix interface in aging and its interaction with the renin-angiotensin system in the aged vasculature. Mech Ageing Dev 2018; 177:66-73. [PMID: 29626500 DOI: 10.1016/j.mad.2018.04.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/22/2018] [Accepted: 04/03/2018] [Indexed: 12/11/2022]
Abstract
The extracellular matrix (ECM) is an intricate network that provides structural and anchoring support to cells in order to stabilize cell morphology and tissue architecture. The ECM also controls many aspects of the cell's dynamic behavior and fate through its ongoing, bidirectional interaction with cells. These interactions between the cell and components of the surrounding ECM are implicated in several biological processes, including development and adult tissue repair in response to injury, throughout the lifespan of multiple species. The present review gives an overview of the growing evidence that cell-matrix interactions play a pivotal role in the aging process. The focus of the first part of the article is on recent studies using cell-derived decellularized ECM, which strongly suggest that age-related changes in the ECM induce cellular senescence, a well-recognized hallmark of aging. This is followed by a review of findings from genetic studies indicating that changes in genes involved in cell-ECM adhesion and matrix-mediated intracellular signaling cascades affect longevity. Finally, mention is made of novel data proposing an intricate interplay between cell-matrix interactions and the renin-angiotensin system that may have a significant impact on mammalian arterial stiffness with age.
Collapse
Affiliation(s)
- Maria De Luca
- Department of Nutrition Sciences, University of Alabama at Birmingham, Webb 451-1720 2nd Ave S, Birmingham, AL, 35294-3360, USA.
| |
Collapse
|
39
|
Yaribeygi H, Farrokhi FR, Rezaee R, Sahebkar A. Oxidative stress induces renal failure: A review of possible molecular pathways. J Cell Biochem 2018; 119:2990-2998. [DOI: 10.1002/jcb.26450] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Accepted: 10/17/2017] [Indexed: 01/04/2023]
Affiliation(s)
- Habib Yaribeygi
- Health Research CenterBaqiyatallah University of Medical SciencesTehranIran
- Chronic Kidney Diseases Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Farin R. Farrokhi
- Chronic Kidney Diseases Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Ramin Rezaee
- Clinical Research Unit, Faculty of MedicineMashhad University of Medical SciencesMashhadIran
| | - Amirhossein Sahebkar
- Biotechnology Research CenterInstitute of Pharmaceutical TechnologyMashhad University of Medical SciencesMashhadIran
- School of PharmacyMashhad University of Medical SciencesMashhadIran
| |
Collapse
|
40
|
Xu XJ, Zhao WB, Feng SB, Sun C, Chen Q, Ni B, Hu HY. Celastrol alleviates angiotensin II‑mediated vascular smooth muscle cell senescence via induction of autophagy. Mol Med Rep 2017; 16:7657-7664. [PMID: 28944849 DOI: 10.3892/mmr.2017.7533] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 07/28/2017] [Indexed: 12/09/2022] Open
Abstract
Reactive oxygen species (ROS) production has been implicated in the promotion of cellular senescence. Celastrol, a quinone methide triterpenoid isolated from the Celastraceae family, exerts antioxidant effects and enhances autophagy in various cell types. Since autophagy serves an important role in regulating ROS, it was hypothesized that the antioxidant effect of celastrol is via enhanced autophagy, thus inhibiting cell senescence. Therefore, the present study used a Senescence β‑Galactosidase Staining kit, western blot analysis and cell cycle analysis to investigate whether celastrol alleviates angiotensin (Ang) II‑induced cellular senescence by upregulating autophagy in vascular smooth muscle cells (VSMCs). The results demonstrated that celastrol reduced Ang II‑induced senescence of VSMCs. Ang II‑induced generation of ROS and the subsequent VSMC senescence were counteracted by pretreatment with celastrol, determined by a ROS assay kit. Celastrol significantly upregulated VSMC autophagy, which reduced intracellular ROS and the subsequent cellular senescence induced by Ang II. Furthermore, celastrol markedly suppressed activity of the mechanistic target of rapamycin signaling pathway in VSMCs. In conclusion, the present study demonstrated that celastrol counteracts VSMC senescence probably by reducing ROS production via activation of autophagy, which may hold promise for the prevention and treatment of aging‑associated cardiovascular disorders such as atherosclerosis.
Collapse
Affiliation(s)
- Xian-Jie Xu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Wei-Bo Zhao
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Shi-Bin Feng
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Cheng Sun
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Qiang Chen
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| | - Bing Ni
- Department of Pathophysiology and High Altitude Pathology, Third Military Medical University, Chongqing 400038, P.R. China
| | - Hou-Yuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing 400038, P.R. China
| |
Collapse
|
41
|
Anwar M, Ahmad S, Akhtar R, Mahmood A, Mahmood S. Antioxidant Supplementation: A Linchpin in Radiation-Induced Enteritis. Technol Cancer Res Treat 2017; 16:676-691. [PMID: 28532242 PMCID: PMC5762044 DOI: 10.1177/1533034617707598] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Radiation enteritis is one of the most feared complications of abdominal and pelvic regions. Thus, radiation to abdominal or pelvic malignancies unavoidably injures the intestine. Because of rapid cell turnover, the intestine is highly sensitive to radiation injury, which is the limiting factor in the permissible dosage of irradiation. Bowel injuries such as fistulas, strictures, and chronic malabsorption are potentially life-threatening complications and have an impact on patient quality of life. The incidence of radiation enteritis is increasing because of the current trend of combined chemotherapy and radiation. The consequences of radiation damage to the intestine may result in considerable morbidity and even mortality. The observed effects of ionizing radiation are mediated mainly by oxygen-free radicals that are generated by its action on water and are involved in several steps of signal transduction cascade, leading to apoptosis. The oxyradicals also induce DNA strand breaks and protein oxidation. An important line of defense against free radical damage is the presence of antioxidants. Therefore, administration of antioxidants may ameliorate the radiation-induced damage to the intestine.
Collapse
Affiliation(s)
- Mumtaz Anwar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shabeer Ahmad
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Reyhan Akhtar
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Akhtar Mahmood
- Department of Biochemistry, Panjab University, Chandigarh, India
| | - Safrun Mahmood
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|