1
|
Zhang X, Li B, Yan Y, Sun F, Zhang S, Wang M, Liu H. AT1R autoantibody promotes phenotypic transition of smooth muscle cells by activating AT1R-OAS2. Biochem Pharmacol 2024; 219:115977. [PMID: 38092283 DOI: 10.1016/j.bcp.2023.115977] [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: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023]
Abstract
Phenotypic transition of vascular smooth muscle cells (VSMCs) is an early event in the onset and progression of several cardiovascular diseases. As an important mediator of the renin-angiotensin system (RAS), activation of the angiotensin II type 1 receptor (AT1R) induces phenotypic transition of VSMCs. AT1R autoantibodies (AT1-AAs), which are agonistic autoantibodies of AT1R, have been detected in the sera of patients with a variety of cardiovascular diseases associated with phenotypic transition. However, the effect of AT1-AA on phenotypic transition is currently unknown. In this study, AT1-AA-positive rat model was established by active immunization to detect markers of VSMCs phenotypic transition. The results showed that AT1-AA-positive rats showed phenotypic transition of VSMCs, which was evidenced by the decrease of contractile markers, while the increase of synthetic markers in the thoracic aorta. However, in AT1-AA-positive AT1R knockout rats, the phenotypic transition-related proteins were not altered. In vitro, after stimulating human aortic smooth muscle cells with AT1-AA for 48 h, 2'-5' oligoadenylate synthase 2 (OAS2) was identified as the key differentially expressed gene by RNA sequencing and bioinformatics analysis. Furthermore, high expression of OAS2 was found in aorta of AT1-AA-positive rats; knockdown of OAS2 by siRNA can reverse the phenotypic transition of VSMCs induced by AT1-AA. In summary, this study suggests that AT1-AA can promote phenotypic transition of VSMCs through AT1R-OAS2 pathway, and OAS2 might serve as a potential therapeutic target to prevent pathological phenotypic transition of smooth muscle cells.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Bingjie Li
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Yingshuo Yan
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Fei Sun
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China
| | - Meili Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
2
|
Allué-Guardia A, Torrelles JB, Sigal A. Tuberculosis and COVID-19 in the elderly: factors driving a higher burden of disease. Front Immunol 2023; 14:1250198. [PMID: 37841265 PMCID: PMC10569613 DOI: 10.3389/fimmu.2023.1250198] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Mycobacterium tuberculosis (M.tb) and SARS-CoV-2 are both infections that can lead to severe disease in the lower lung. However, these two infections are caused by very different pathogens (Mycobacterium vs. virus), they have different mechanisms of pathogenesis and immune response, and differ in how long the infection lasts. Despite the differences, SARS-CoV-2 and M.tb share a common feature, which is also frequently observed in other respiratory infections: the burden of disease in the elderly is greater. Here, we discuss possible reasons for the higher burden in older adults, including the effect of co-morbidities, deterioration of the lung environment, auto-immunity, and a reduced antibody response. While the answer is likely to be multifactorial, understanding the main drivers across different infections may allow us to design broader interventions that increase the health-span of older people.
Collapse
Affiliation(s)
- Anna Allué-Guardia
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Jordi B. Torrelles
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX, United States
- International Center for the Advancement of Research and Education (I•CARE), Texas Biomedical Research Institute, San Antonio, TX, United States
| | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
3
|
Shiwakoti S, Gong D, Sharma K, Kang KW, Schini-Kerth VB, Kim HJ, Ko JY, Oak MH. γ-Oryzanol ameliorates fine dust-induced premature endothelial senescence and dysfunction via attenuating oxidative stress. Food Chem Toxicol 2023; 179:113981. [PMID: 37549806 DOI: 10.1016/j.fct.2023.113981] [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: 05/11/2023] [Revised: 07/24/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Various cardiovascular diseases are associated with endothelial senescence, and a recent study showed that fine dust (FD)-induced premature endothelial senescence and dysfunction is associated with increased oxidative stress. The aim of the present study was to investigate protective effect of rice bran extract (RBE) and its major component of γ-Oryzanol (γ-Ory) against FD-induced premature endothelial senescence. Porcine coronary artery endothelial cells (PCAECs) were treated with FD alone or with RBE or γ-Ory. Senescence-associated β-galactosidase (SA-β-gal) activity, expression of cell cycle regulatory proteins, and oxidative stress levels were evaluated. The results indicated that SA-β-gal activity in the FD-treated PCAECs was attenuated by RBE and γ-Ory. Additionally, γ-Ory inhibited FD-induced cell cycle arrest, restored cell proliferation, and reduced the expression of cell cycle regulatory proteins. γ-Ory also inhibited oxidative stress and prevented senescence-associated NADPH oxidase and LAS activity in FD-exposed ECs suggesting that γ-Ory could protect against FD-induced ECs senescence and dysfunction.
Collapse
Affiliation(s)
- Saugat Shiwakoti
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Dalseong Gong
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea; Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260, INSERM (French National Institute of Health and Medical Research), University of Strasbourg, 67000, Strasbourg, France
| | - Kushal Sharma
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Ki-Woon Kang
- Division of Cardiology, College of Medicine, Heart Reasearch Institute and Biomedical Research Institute, Chung-Ang University Hospital, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Valérie B Schini-Kerth
- Regenerative Nanomedicine, Faculty of Pharmacy, UMR 1260, INSERM (French National Institute of Health and Medical Research), University of Strasbourg, 67000, Strasbourg, France
| | - Hyun Jung Kim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea
| | - Ju-Young Ko
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea.
| | - Min-Ho Oak
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, Jeonnam, 58554, Republic of Korea.
| |
Collapse
|
4
|
Wang M, Yin X, Li S, Zhang X, Yi M, He C, Li X, Wang W, Zhang S, Liu H. Large‐Conductance Calcium‐Activated Potassium Channel Opener, NS1619, Protects Against Mesenteric Artery Remodeling Induced by Agonistic Autoantibodies Against the Angiotensin II Type 1 Receptor. J Am Heart Assoc 2022; 11:e024046. [PMID: 35156422 PMCID: PMC9245824 DOI: 10.1161/jaha.121.024046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background Agonistic autoantibodies against the angiotensin II type 1 receptor (AT1‐AAs) extensively exist in patients with hypertensive diseases and have been demonstrated to play crucial roles in the pathophysiological process of vascular remodeling. However, the treatment options are limited. The large‐conductance calcium‐activated potassium (BK) channel is a critical regulator and potential therapeutic target of vascular tone and architecture. We have previously observed that AT1‐AAs have an inhibitory effect on BK channels. However, whether BK channel dysfunction is involved in AT1‐AAs‐induced vascular remodeling and the therapeutic effect of BK channel opener is unclear. Methods and Results In our study, mesenteric arteries from AT1‐AAs‐positive rats exhibited increased wall thickness, narrowing of the arteriolar lumen, and increased collagen accumulation. Patch clamp test results showed that the voltage sensitivity of BK channel declined in mesenteric arteriolar smooth muscle cells from AT1‐AAs‐positive rats. Experiments with freshly isolated mesenteric arteriolar smooth muscle cells showed that AT1‐AAs reduced the opening probability, open levels, open dwell time, and calcium sensitivity of BK channel. Experiments with HEK293T cells transfected with GFP‐ZERO‐BK α‐subunit plasmids suggested a BK channel α‐subunit‐dependent mechanism. BK channel α‐subunit deficient, namely KCNMA1−/− rats showed a phenotype of mesenteric artery remodeling. The administration of NS1619, a specific BK channel opener targeting the α‐subunit, reversed the phenotypic transition and migration induced by AT1‐AAs in cultured mesenteric arteriolar smooth muscle cells. Finally, perfusion of NS1619 significantly relieved the pathological effects induced by AT1‐AAs in vivo. Conclusions In summary, we provide compelling evidence that BK channel α‐subunit dysfunction mediates AT1‐AAs‐induced mesenteric artery remodeling. Preservation of BK channel activity may serve as a potential strategy for the treatment of AT1‐AAs‐induced maladaptive resistance artery remodeling.
Collapse
Affiliation(s)
- Meili Wang
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular DiseaseCapital Medical University Beijing China
| | - Xiaochen Yin
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Shuanglei Li
- Division of Adult Cardiac Surgery Department of Cardiology The Sixth Medical CenterChinese PLA General Hospital Beijing China
| | - Xi Zhang
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Ming Yi
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Chunyu He
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Xiaoyue Li
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
| | - Wei Wang
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular DiseaseCapital Medical University Beijing China
| | - Suli Zhang
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular DiseaseCapital Medical University Beijing China
| | - Huirong Liu
- Department of Physiology and Pathophysiology School of Basic Medical Sciences Capital Medical University Beijing China
- Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular DiseaseCapital Medical University Beijing China
| |
Collapse
|
5
|
Wu XW, Li G, Cheng XB, Wang M, Wang LL, Wang HH, Yang JY, Hu XJ. Association of Angiotensin II Type 1 Receptor Agonistic Autoantibodies With Outcomes in Patients With Acute Aortic Dissection. JAMA Netw Open 2021; 4:e2127587. [PMID: 34596673 PMCID: PMC8486983 DOI: 10.1001/jamanetworkopen.2021.27587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
IMPORTANCE Angiotensin II is significantly associated with the pathogenesis of acute aortic dissection. Angiotensin II type 1 receptor agonistic autoantibodies (AT1-AAs) can mimic the effect of angiotensin II. OBJECTIVE To investigate the association between AT1-AAs and all-cause and cause-specific mortality risk in patients with acute aortic dissection. DESIGN, SETTING, AND PARTICIPANTS A total of 662 patients with clinically suspected aortic dissection from 3 medical centers in Wuhan, China, were enrolled in this cohort study from August 1, 2014, to July 31, 2016. Of these, 315 patients were included in the 3-year follow-up study. Follow-up was mainly performed via telephone interviews and outpatient clinic visits. Data analysis was conducted from March 1 to May 31, 2020. MAIN OUTCOMES AND MEASURES The primary outcomes of interest were all-cause mortality, death due to aortic dissection, and late aortic-related adverse events. RESULTS The full study cohort included 315 patients with AAD (mean [SD] age, 56.2 [12.7] years; 230 men [73.0%]). Ninety-two patients (29.2%) were positive for AT1-AAs. The mortality of AT1-AA-positive patients was significantly higher than that of AT1-AA-negative patients (40 [43.5%] vs 37 [16.6%]; P < .001). The mortality risk in AT1-AA-positive patients was always significantly higher than that in AT1-AA-negative patients in patients with both type A and type B dissection. Multivariable analysis showed that the risk of AT1-AA-positive patients for type A dissection was significantly higher than that of AT1-AA-negative patients (odds ratio [OR], 1.88; 95% CI, 1.12-3.13; P = .02). The Cox proportional hazards regression model showed a significant increase of all-cause mortality risk (OR, 2.27; 95% CI, 1.44-3.57; P < .001) and late aortic-related adverse events (OR, 1.58; 95% CI, 1.06-2.36; P = .03) among AT1-AA-positive patients during the follow-up period compared with AT1-AA-negative patients. CONCLUSIONS AND RELEVANCE This cohort study first detected AT1-AAs in patients with acute aortic dissection. The presence of AT1-AAs was associated with significantly higher all-cause and cause-specific mortality during a follow-up period of 3 years. The antibodies may be a risk factor for aortic dissection.
Collapse
Affiliation(s)
- Xiao-wei Wu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Li
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
- Hubei Province Academy of Traditional Chinese Medicine, Wuhan, China
| | - Xiao-bin Cheng
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Min Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Liu-lin Wang
- Emergency Department, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China
| | - Hai-hao Wang
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-ye Yang
- Department of Cardiothoracic and Vascular Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-jian Hu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Freitag H, Szklarski M, Lorenz S, Sotzny F, Bauer S, Philippe A, Kedor C, Grabowski P, Lange T, Riemekasten G, Heidecke H, Scheibenbogen C. Autoantibodies to Vasoregulative G-Protein-Coupled Receptors Correlate with Symptom Severity, Autonomic Dysfunction and Disability in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. J Clin Med 2021; 10:3675. [PMID: 34441971 PMCID: PMC8397061 DOI: 10.3390/jcm10163675] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS) is an acquired complex disease with patients suffering from the cardinal symptoms of fatigue, post-exertional malaise (PEM), cognitive impairment, pain and autonomous dysfunction. ME/CFS is triggered by an infection in the majority of patients. Initial evidence for a potential role of natural regulatory autoantibodies (AAB) to beta-adrenergic (AdR) and muscarinic acetylcholine receptors (M-AChR) in ME/CFS patients comes from a few studies. METHODS Here, we analyzed the correlations of symptom severity with levels of AAB to vasoregulative AdR, AChR and Endothelin-1 type A and B (ETA/B) and Angiotensin II type 1 (AT1) receptor in a Berlin cohort of ME/CFS patients (n = 116) by ELISA. The severity of disease, symptoms and autonomic dysfunction were assessed by questionnaires. RESULTS We found levels of most AABs significantly correlated with key symptoms of fatigue and muscle pain in patients with infection-triggered onset. The severity of cognitive impairment correlated with AT1-R- and ETA-R-AAB and severity of gastrointestinal symptoms with alpha1/2-AdR-AAB. In contrast, the patients with non-infection-triggered ME/CFS showed fewer and other correlations. CONCLUSION Correlations of specific AAB against G-protein-coupled receptors (GPCR) with symptoms provide evidence for a role of these AAB or respective receptor pathways in disease pathomechanism.
Collapse
Affiliation(s)
- Helma Freitag
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Marvin Szklarski
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Sebastian Lorenz
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Franziska Sotzny
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Sandra Bauer
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Aurélie Philippe
- Department of Nephrology and Critical Care Medicine, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany;
| | - Claudia Kedor
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Patricia Grabowski
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
| | - Tanja Lange
- Department of Rheumatology and Clinical Immunology, University of Lübeck, 23538 Lübeck, Germany; (T.L.); (G.R.)
| | - Gabriela Riemekasten
- Department of Rheumatology and Clinical Immunology, University of Lübeck, 23538 Lübeck, Germany; (T.L.); (G.R.)
| | | | - Carmen Scheibenbogen
- Institute of Medical Immunology, Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (M.S.); (S.L.); (F.S.); (S.B.); (C.K.); (P.G.); (C.S.)
- Berlin Institute of Health Center for Regenerative Therapies (BCRT), Charité—Universitätsmedizin Berlin, 10117 Berlin, Germany
| |
Collapse
|
7
|
He C, Li X, Wang M, Zhang S, Liu H. Deletion of BK channels decreased skeletal and cardiac muscle function but increased smooth muscle contraction in rats. Biochem Biophys Res Commun 2021; 570:8-14. [PMID: 34271438 DOI: 10.1016/j.bbrc.2021.07.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/07/2021] [Indexed: 12/28/2022]
Abstract
Large conductance calcium-activated potassium channel (BK channel) is widely expressed in skeletal muscle, myocardium, smooth muscle and other muscle tissues. Mutation, abnormal expression and altered activity of BK channel are linked to muscle-related diseases such as dyskinesia, epilepsy and erectile dysfunction. In order to compare the effects of BK channel on different muscle tissues, we constructed BK channel gene knockout rats (BK-/- rats). HE staining, open field and grip strength tests, ultrasound, blood pressure measurement and vascular tension test were utilized to explore the effects of BK channel deletion on the structure and function changes in skeletal muscle, myocardium, and vascular smooth muscle (VSM). It was found that compared with wild-type rats, the BK-/- rats showed decreased skeletal muscle fiber area, grip, movement distance and speed at 2 and 12 months of ages. At heart, the muscle fiber area, cardiac systolic/diastolic function and heart rate decreased in BK-/- rats. The wall of the left ventricle became thin. However, the vascular wall of BK-/- rats thickened, the pulse wave velocity was increased, and the VSM contraction was enhanced. Unexpectedly, both systolic and diastolic blood pressure were reduced in BK-/- rats, while pulse pressure difference was increased. These results suggest that BK channel may have different effects on different types of muscle tissue, and it should be noted that different parts of muscle tissue may have different effects when BK channel-related drugs are used.
Collapse
Affiliation(s)
- Chunyu He
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Beijing, 100069, China
| | - Xiaoyue Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Beijing, 100069, China
| | - Meili Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Beijing, 100069, China
| | - Suli Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Beijing, 100069, China.
| | - Huirong Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Beijing, 100069, China.
| |
Collapse
|
8
|
Liu Z, Xiao M, Du Z, Li M, Guo H, Yao M, Wan X, Xie Z. Dietary supplementation of Huangshan Maofeng green tea preventing hypertension of older C57BL/6 mice induced by desoxycorticosterone acetate and salt. J Nutr Biochem 2021; 88:108530. [PMID: 33080347 DOI: 10.1016/j.jnutbio.2020.108530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 08/26/2020] [Accepted: 10/14/2020] [Indexed: 01/12/2023]
Abstract
Senile hypertension affects the life quality of aged population. Dietary intervention plays a pivotal role in the prevention of hypertension. There are few reports concerning the effects and mechanisms of green tea supplementation preventing age related hypertension. The current study investigated the effect and mechanism of dietary supplement of Huangshan Maofeng green tea (HSMF) on prevention of hypertension induced by deoxycorticosterone acetate (DOCA) and salt in old C57BL/6 mice. Our results showed that HSMF dose-dependently prevented the increase of systolic blood pressure and diastolic blood pressure induced by DOCA plus salt (DS) at 51-week-old mice. And HSMF significantly reduced the agonists' stimulated contraction of mesenteric arteries isolated from the old mice. The expression of vasoconstrictor genes and inflammatory cytokines in aorta were suppressed observably by HSMF supplementation compared with DS group. The protein expression of PKCα in the aorta was dose-dependently decreased by HSMF compared to DS group. The phosphorylation level of MYPT1, CPI-17and MLC20 was also restrained by HSMF in the aorta. Furthermore, HSMF protected kidney by maintaining integrity of glomeruli and tubules and remarkably decreased the NGAL level in plasma. HSMF also suppressed the kidney inflammation by decreasing inflammatory cytokines expression and the macrophage infiltration. Our results proved that dietary supplement of HSMF remarkably improved the vascular functions and protected kidney injury, and thus prevented hypertension induced by DS in older C57BL/6 mice. Our data indicated that the dietary supplement of HSMF may potentially be used as a food additive for preventing hypertension for aged people.
Collapse
Affiliation(s)
- Zenghui Liu
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China; Anhui Academy of Medical Science, Hefei, China
| | - Mengchao Xiao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Zhaofeng Du
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China; School of Biology and Food Engineering, Fuyang Normal University, Fuyang, China
| | - Mengwan Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Huimin Guo
- Center for Biotechnology, Anhui Agricultural University, Hefei, China
| | - Min Yao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China
| | - Xiaochun Wan
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| | - Zhongwen Xie
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea and Food Sciences and Technology, Anhui Agricultural University, Hefei, China.
| |
Collapse
|
9
|
Shiwakoti S, Adhikari D, Lee JP, Kang KW, Lee IS, Kim HJ, Oak MH. Prevention of Fine Dust-Induced Vascular Senescence by Humulus lupulus Extract and Its Major Bioactive Compounds. Antioxidants (Basel) 2020; 9:E1243. [PMID: 33297587 PMCID: PMC7762380 DOI: 10.3390/antiox9121243] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/30/2020] [Accepted: 12/03/2020] [Indexed: 12/23/2022] Open
Abstract
Both short- and long-term exposure to fine dust (FD) from air pollution has been linked to various cardiovascular diseases (CVDs). Endothelial cell (EC) senescence is an important risk factor for CVDs, and recent evidence suggests that FD-induced premature EC senescence increases oxidative stress levels. Hop plant (Humulus lupulus) is a very rich source of polyphenols known to have nutritional and therapeutic properties, including antioxidant behavior. The aims of this study were to evaluate whether Humulus lupulus extract prevents FD-induced vascular senescence and dysfunction and, if so, to characterize the underlying mechanisms and active components. Porcine coronary arteries and endothelial cells were treated with FD in the presence or absence of hop extract (HOP), and the senescence-associated-beta galactosidase (SA-β-gal) activity, cell-cycle progression, expression of senescence markers, oxidative stress level, and vascular function were evaluated. Results indicated that HOP inhibited FD-induced SA-β-gal activity, cell-cycle arrest, and oxidative stress, suggesting that HOP prevents premature induction of senescence by FD. HOP also ameliorated FD-induced vascular dysfunction. Additionally, xanthohumol (XN) and isoxanthohumol (IX) were found to produce the protective effects of HOP. Treatment with HOP and its primary active components XN and IX downregulated the expression of p22phox, p53, and angiotensin type 1 receptor, which all are known FD-induced redox-sensitive EC senescence inducers. Taken together, HOP and its active components protect against FD-induced endothelial senescence most likely via antioxidant activity and may be a potential therapeutic agent for preventing and/or treating air-pollution-associated CVDs.
Collapse
Affiliation(s)
- Saugat Shiwakoti
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Deepak Adhikari
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Jeong Pyo Lee
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Ki-Woon Kang
- Division of Cardiology, Eulji University Hospital, Eulji University School of Medicine, Daejeon 34824, Korea;
| | - Ik-Soo Lee
- College of Pharmacy, Chonnam National University, Gwangju 61186, Korea;
| | - Hyun Jung Kim
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| | - Min-Ho Oak
- College of Pharmacy, Mokpo National University, Jeonnam, Muan-gun 58554, Korea; (S.S.); (D.A.); (J.P.L.)
| |
Collapse
|
10
|
Zhang Y, Song Z, Huang S, Zhu L, Liu T, Shu H, Wang L, Huang Y, Chen Y. Aloe emodin relieves Ang II-induced endothelial junction dysfunction via promoting ubiquitination mediated NLRP3 inflammasome inactivation. J Leukoc Biol 2020; 108:1735-1746. [PMID: 32573820 PMCID: PMC7754316 DOI: 10.1002/jlb.3ma0520-582r] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 01/07/2023] Open
Abstract
Recent studies have revealed that aloe emodin (AE), a natural compound from the root and rhizome of Rheum palmatum L., exhibits significant pharmacologic activities. However, the pharmacologic relevance of the compound, particularly for cardiovascular disease, remains largely unknown. Here, we hypothesized that AE could improve endothelial junction dysfunction through inhibiting the activation of NOD-like receptor family pyrin domain containing-3 (NLRP3) inflammasome regulated by NLRP3 ubiquitination, and ultimately prevent cardiovascular disease. In vivo, we used confocal microscopy to study the expression of tight junction proteins zonula occludens-1/2 (ZO-1/2) and the formation of NLRP3 inflammasome in coronary arteries of hypertension. And the experimental serum was used to detect the activation of NLRP3 inflammasome by ELISA assay. We found that AE could restore the expression of the endothelial connective proteins ZO-1/2 and decrease the release of high mobility group box1 (HMGB1), and also inhibited the formation and activation of NLRP3 inflammasome. Similarly, in vitro, our findings demonstrated that AE could restore the expression of the tight junction proteins ZO-1/2 and decrease monolayer cell permeability that related to endothelial function after stimulation by angiotensin II (Ang II) in microvascular endothelial cells (MECs). We also demonstrated that AE could inhibit Ang II-induced NLRP3 inflammasome formation and activation, which were regulated by NLRP3 ubiquitination in MECs, as shown by fluorescence confocal microscopy and Western blot. Together with these changes, we revealed a new protection mechanism of AE that inhibited NLRP3 inflammasome activation and decreased the release of HMGB1 by promoting NLRP3 ubiquitination. Our findings implicated that AE exhibited immense potential and specific therapeutic value in hypertension-related cardiovascular disease in the early stage and the development of innovative drugs.
Collapse
Affiliation(s)
- Yi Zhang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, Guangzhou, China
| | - Ziqing Song
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, Guangzhou, China
| | - Shan Huang
- Department of Stomatology, The First Affiliated Hospital, The School of Dental Medicine, Jinan University, Guangzhou, China
| | - Li Zhu
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, Guangzhou, China
| | - Tianyi Liu
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, Guangzhou, China
| | - Hongyan Shu
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, Guangzhou, China
| | - Lei Wang
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, Guangzhou, China
| | - Yi Huang
- Department of Stomatology, The First Affiliated Hospital, The School of Dental Medicine, Jinan University, Guangzhou, China
| | - Yang Chen
- School of Pharmaceutical, Guangzhou University of Chinese Medicine, Guangzhou University Town, Guangzhou, China
| |
Collapse
|
11
|
Jain A, Shah H, Simonsick EM, Metter EJ, Mangold L, Humphreys E, Partin A, Fedarko NS. Angiotensin receptor autoantibodies as exposures that modify disease progression: Cross sectional, longitudinal and in vitro studies of prostate cancer. J Transl Autoimmun 2019; 2:100008. [PMID: 31930191 PMCID: PMC6953913 DOI: 10.1016/j.jtauto.2019.100008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Revised: 07/28/2019] [Accepted: 07/29/2019] [Indexed: 12/23/2022] Open
Abstract
Circulating angiotensin type I receptor (AT1R) agonistic autoantibodies (AT1RaAbs) that bind and chronically activate the receptor have been associated with a number of diseases suggesting that while the autoantibodies are not necessarily causative they may promote disease progression. The prostate has a local renin angiotensin system. The current study examines associations between AT1RaAbs and prostate cancer (PCA), disease-free survival (DFS), overall survival (OS) and AT1RaAb effects on PCA cell phenotype. In a cross-sectional set of serum obtained from 151 men diagnosed with PCA, nonmalignant prostate disease or no disease, higher serum AT1RaAb levels were associated with PCA and non-organ confined PCA. The odds ratio for PCA was 6.3 (95% confidence interval 2.2 to 18) for a positive 1:1600 titer and 18 (95% confidence interval 6.9 to 45) at AT1RaAb levels > 1.04 μg/ml, (p < 0.0001). In a longitudinal set of pre-diagnosis samples from 109 men, DFS hazard ratios of 2.2 (95% confidence interval 1.4 to 3.5) and 1.6 (95% confidence interval 1.0 to 2.5) for most proximal to diagnosis and most distal to diagnosis samples, respectively, were found for high versus low AT1RaAb groups. Hazard ratios for OS in most proximal and distal samples were 2.4 (95% confidence interval 1.6 to 3.6) and 1.8 (95% confidence interval 1.1 to 2.8), respectively. Accelerated failure modeling of survival indicated that a 1 μg/ml increase in AT1RaAb levels was associated with a reduction of DFS and OS by 20% at the most proximal time point and by 15% at the most distal time points. Adjusting for age, did not affect the association with DFS in proximal samples but changed distal time point DFS and OS to a 10% decrease for every 1 μg/ml increase in AT1RaAb. Additional adjustments for body mass index, systolic blood pressure and prostate-specific antigen did not appreciably alter these associations. AT1RaAb treatment of PC3, DU145, and LNCaP cells significantly increased the maximal growth rate approximately 2-fold and invasiveness approximately 3-fold. Conclusions: These observations provide evidence supporting AT1RaAbs as exposures that may modify prostate cancer progression and indicate they may be predictive markers for risk stratification.
Collapse
Affiliation(s)
- Alka Jain
- Department of Medicine, Johns Hopkins University, Baltimore, MD, 21224, USA
| | - Haikoo Shah
- Department of Medicine, Johns Hopkins University, Baltimore, MD, 21224, USA
- Department of Medicine, University Hospitals Cleveland Medical Center, Case Western Reserve University, Cleveland, OH, 44106, USA
| | - Eleanor M. Simonsick
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21225, USA
| | - E. Jeffrey Metter
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD, 21225, USA
- Current Address: Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Leslie Mangold
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Elizabeth Humphreys
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Alan Partin
- Department of Urology, James Buchanan Brady Urological Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Neal S. Fedarko
- Department of Medicine, Johns Hopkins University, Baltimore, MD, 21224, USA
| |
Collapse
|