1
|
Fleckner M, Döhmen NK, Salz K, Christophers T, Windolf J, Suschek CV, Oezel L. Exposure of Primary Human Skin Fibroblasts to Carbon Dioxide-Containing Solution Significantly Reduces TGF-β-Induced Myofibroblast Differentiation In Vitro. Int J Mol Sci 2024; 25:13013. [PMID: 39684728 DOI: 10.3390/ijms252313013] [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: 10/30/2024] [Revised: 11/21/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Wound healing as a result of a skin injury involves a series of dynamic physiological processes, leading to wound closure, re-epithelialization, and the remodeling of the extracellular matrix (ECM). The primary scar formed by the new ECM never fully regains the original tissue's strength or flexibility. Moreover, in some cases, due to dysregulated fibroblast activity, proliferation, and differentiation, the normal scarring can be replaced by pathological fibrotic tissue, leading to hypertrophic scars or keloids. These disorders can cause significant physical impairment and psychological stress and represent significant challenges in medical management in the wound-healing process. The present study aimed to investigate the therapeutic effects of exogenously applied carbon dioxide (CO2) on fibroblast behavior, focusing on viability, proliferation, migration, and differentiation to myofibroblasts. We found that CO2 exposure for up to 60 min did not significantly affect fibroblast viability, apoptosis rate, or proliferation and migration capacities. However, a notable finding was the significant reduction in α-smooth muscle actin (α-SMA) protein expression, indicative of myofibroblast differentiation inhibition, following CO2 exposure. This effect was specific to CO2 and concentration as well as time-dependent, with longer exposure durations leading to greater reductions in α-SMA expression. Furthermore, the inhibition of myofibroblast differentiation correlated with a statistically significantly reduced glycolytic and mitochondrial energy metabolism, and as a result, with a reduced ATP synthesis rate. This very noticeable decrease in cellular energy levels seemed to be specific to CO2 exposure and could not be observed in the control cultures using nitrogen (N2)-saturated solutions, indicating a unique and hypoxia-independent effect of CO2 on fibroblast metabolism. These findings suggest that exogenously applied CO2 may possess fibroblast differentiation-reducing properties by modulating fibroblast's energy metabolism and could offer new therapeutic options in the prevention of scar and keloid development.
Collapse
Affiliation(s)
- Maxine Fleckner
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Niklas K Döhmen
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Katharina Salz
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Till Christophers
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Joachim Windolf
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Christoph V Suschek
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| | - Lisa Oezel
- Department for Orthopedics and Trauma Surgery, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, 40225 Dusseldorf, Germany
| |
Collapse
|
2
|
Odanaka Y, Kishi K, Takigiku K, Ashida A, Ozaki N, Ashida A. Microvascular Endothelial Function Assessed Using Peripheral Arterial Tonometry in Adolescents with Repaired Congenital Heart Disease. Pediatr Cardiol 2024; 45:1804-1810. [PMID: 37697168 DOI: 10.1007/s00246-023-03283-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 08/19/2023] [Indexed: 09/13/2023]
Abstract
Atherosclerosis can develop in adult patients with congenital heart disease (CHD) and should be given attention. Endothelial function is well known as a predictor of the development of atherosclerosis but has not been well investigated in patients with repaired CHD. This study aimed to clarify the endothelial function and its relationship with clinical backgrounds and parameters in adolescents with various types of repaired CHDs. Endothelial function was evaluated using peripheral arterial tonometry (PAT). The reactive hyperemia index (RHI) was evaluated and compared between adolescents with repaired CHD and those in the control group. The relationship between the clinical background and parameters was also investigated in patients with repaired CHD. Forty-eight patients with repaired CHD (age 14.0 ± 3.3 years) and 114 healthy volunteers were included in this study. Patients with repaired CHD comprised 16 with repaired non-cyanotic CHD, 14 with repaired tetralogy of Fallot, and 18 who underwent the Fontan procedure. RHI in the repaired CHD group was significantly lower than in the control group. There was no significant correlation between the RHI and blood biochemical markers, such as uric acid, creatine, and brain natriuretic peptide levels. The RHI was significantly higher in patients taking angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs) than in those not taking them. Endothelial function was impaired in adolescents with repaired CHD compared to that in the control group. Microvascular endothelial dysfunction developed even in adolescents with simple non-cyanotic CHD.
Collapse
Affiliation(s)
- Yutaka Odanaka
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan.
| | - Kanta Kishi
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Kiyohiro Takigiku
- Department of Pediatric Cardiology, Nagano Children's Hospital, Nagano, Japan
| | - Atsuko Ashida
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Noriyasu Ozaki
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| | - Akira Ashida
- Department of Pediatrics, Osaka Medical and Pharmaceutical University, 2-7 Daigakumachi, Takatsuki, Osaka, 569-8686, Japan
| |
Collapse
|
3
|
Ajoolabady A, Pratico D, Ren J. Angiotensin II: Role in oxidative stress, endothelial dysfunction, and diseases. Mol Cell Endocrinol 2024; 592:112309. [PMID: 38852657 DOI: 10.1016/j.mce.2024.112309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/11/2024]
Abstract
Angiotensin II (Ang II) is a protein hormone capable of physiologically regulating blood pressure through diverse mechanisms. Ang II is mainly produced by the liver at homeostatic levels. However, excessive production of Ang II is closely associated with a series of pathological events in the body. The endothelial dysfunction is one of these pathological events that can drive vascular anomalies. The excessive exposure of endothelial cells (ECs) to Ang II may induce endothelial dysfunction via diverse mechanisms. One of these mechanisms is Ang II-mediated mitochondrial oxidative stress. In this mini-review, we aimed to discuss the molecular mechanisms of Ang II-mediated endothelial dysfunction through mitochondrial oxidative stress and the protective role of nitric oxide in ECs. Deciphering these mechanisms may disclose novel therapeutic strategies to prevent endothelial dysfunction and associated diseases induced by elevated leves of Ang II in the blood.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai, 200032, China.
| |
Collapse
|
4
|
Abduljabbar MH, Alnemari RM, Almalki AH, Alqarni M, Abdel Rahman MA, Serag A. Continuous Wavelet Transform as a Signal Processing Technique for Spectrofluorimetric Analysis of Rosuvastatin and Losartan: Greenness and Blueness Assessment. LUMINESCENCE 2024; 39:e4928. [PMID: 39428109 DOI: 10.1002/bio.4928] [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: 07/08/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/22/2024]
Abstract
In this study, the application of continuous wavelet transform as a signal processing technique for the spectrofluorimetric determination of rosuvastatin and losartan is investigated. Both rosuvastatin and losartan exhibited native spectrofluorometric signals with severe overlapping peaks, making their simultaneous determination challenging. To address this issue, rbio 2.4 wavelet transformation was employed to obtain zero-crossing points in the synchronous fluorescence spectra of losartan and rosuvastatin at 346 and 408 nm, respectively, making their quantification possible. The developed method was validated according to the ICH guidelines and displayed high accuracy, precision, and specificity. The method exhibited excellent linearity over concentration ranges 0.2-2.0 and 0.25-2.0 μg/mL for losartan and rosuvastatin, respectively. In addition, LOD and LOQ were 0.046 and 0.140 μg/mL for losartan and 0.036 and 0.110 μg/mL for rosuvastatin, respectively, indicating the high sensitivity of the developed method. Moreover, greenness and blueness assessments were carried, revealing a high AGREE score of 0.71 and BAGI score of 77.5 for the developed method, making it a promising greener alternative for the reported chromatographic methods. Finally, the developed method was applied to the determination of rosuvastatin and losartan in pharmaceutical formulations, posing it as a powerful greener alternative in quality control laboratories.
Collapse
Affiliation(s)
- Maram H Abduljabbar
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Reem M Alnemari
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Atiah H Almalki
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
- Addiction and Neuroscience Research Unit, Health Science Campus, Taif University, Taif, Saudi Arabia
| | - Mohammed Alqarni
- Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mona A Abdel Rahman
- Analytical Chemistry Department, Faculty of Pharmacy, October 6 University, Giza, Egypt
| | - Ahmed Serag
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
5
|
Yin XM, Song YY, Jiang WY, Zhang HT, Chen JW, Murao K, Han MX, Sun WP, Zhang GX. Mitochondrial K ATP channel-mediated autophagy contributes to angiotensin II-induced vascular dysfunction in mice. Nutr Metab Cardiovasc Dis 2024; 34:1571-1580. [PMID: 38418351 DOI: 10.1016/j.numecd.2024.01.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 10/28/2023] [Accepted: 01/15/2024] [Indexed: 03/01/2024]
Abstract
BACKGROUND AND AIM The present study aimed to investigate whether the mitochondrial KATP channel contributes to angiotensin II (Ang II)-induced vascular dysfunction, the development of hypertension, and atherosclerosis. METHODS AND RESULTS ApoE (-/-) mice fed a high-fat diet were chronically infused with Ang II for eight weeks and concomitantly treated with losartan (ARB), apocynin, or 5-hydroxy decanoate (5-HD), or 3-methyladenine (3-MA). Systolic blood pressure was measured, and pathological changes of aortic or liver tissue were observed. Nitric oxide (NO), superoxide dismutase 2 (SOD2) levels and vasorelaxation rate were measured, and protein and mRNA expressions were examined by western blot and RT-PCR. Ang II-induced development of hypertension was suppressed not only by ARB, and apocynin but also by 5-HD or 3-MA. Ang II infusion decreased aortic NO production and relaxation, as well as SOD2 activity in liver, which were improved by all treatments. In addition, Ang II-induced activation of autophagy was suppressed by 5-HD in aortic tissue, furthermore, Ang II increases the atherosclerotic index in plasma and exacerbates the development of atherosclerosis by increases of fat deposition in the aorta and liver. Lipid metabolism-related mRNA expressions (LXR-α, LDLR, SRBI, Acca, and FASN) were changed by Ang II. Similarly, not only ARB, and apocynin, but also 5-HD and 3-MA suppressed Ang II-induced these changes. CONCLUSIONS Our present findings evidence that mitochondrial KATP channel-mediated autophagy contributes to Ang II-induced vascular dysfunction, development of hypertension, and atherosclerosis.
Collapse
Affiliation(s)
- Xue-Min Yin
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Yi-Yi Song
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Wen-Yi Jiang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Hao-Tian Zhang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China
| | - Jing-Wei Chen
- Department of Internal Medicine, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 18 Yang-Su Road, Suzhou 215003, PR China
| | - Koji Murao
- Department of Endocrine and Metabolism, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe Miki-cho, Kita-gun, Kagawa, 761-0793, Japan
| | - Meng-Xiao Han
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China.
| | - Wan-Ping Sun
- Laboratory of Molecular Diagnostics, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China.
| | - Guo-Xing Zhang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China; Suzhou Key Laboratory of Drug Research for Prevention and Treatment of Hyperlipidemic Diseases, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou 215123, PR China.
| |
Collapse
|
6
|
Abdelnaby AE, Trebak M. Store-Operated Ca 2+ Entry in Fibrosis and Tissue Remodeling. CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2024; 7:25152564241291374. [PMID: 39659877 PMCID: PMC11629433 DOI: 10.1177/25152564241291374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/29/2024] [Accepted: 09/27/2024] [Indexed: 12/12/2024]
Abstract
Fibrosis is a pathological condition characterized by excessive tissue deposition of extracellular matrix (ECM) components, leading to scarring and impaired function across multiple organ systems. This complex process is mediated by a dynamic interplay between cell types, including myofibroblasts, fibroblasts, immune cells, epithelial cells, and endothelial cells, each contributing distinctively through various signaling pathways. Critical to the regulatory mechanisms involved in fibrosis is store-operated calcium entry (SOCE), a calcium entry pathway into the cytosol active at the endoplasmic reticulum-plasma membrane contact sites and common to all cells. This review addresses the multifactorial nature of fibrosis with a focus on the pivotal roles of different cell types. We highlight the essential functions of myofibroblasts in ECM production, the transformation of fibroblasts, and the participation of immune cells in modulating the fibrotic landscape. We emphasize the contributions of SOCE in these different cell types to fibrosis, by exploring the involvement of SOCE in cellular functions such as proliferation, migration, secretion, and inflammatory responses. The examination of the cellular and molecular mechanisms of fibrosis and the role of SOCE in these mechanisms offers the potential of targeting SOCE as a therapeutic strategy for mitigating or reversing fibrosis.
Collapse
Affiliation(s)
- Ahmed Emam Abdelnaby
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed Trebak
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Allbritton-King JD, García-Cardeña G. Endothelial cell dysfunction in cardiac disease: driver or consequence? Front Cell Dev Biol 2023; 11:1278166. [PMID: 37965580 PMCID: PMC10642230 DOI: 10.3389/fcell.2023.1278166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/09/2023] [Indexed: 11/16/2023] Open
Abstract
The vascular endothelium is a multifunctional cellular system which directly influences blood components and cells within the vessel wall in a given tissue. Importantly, this cellular interface undergoes critical phenotypic changes in response to various biochemical and hemodynamic stimuli, driving several developmental and pathophysiological processes. Multiple studies have indicated a central role of the endothelium in the initiation, progression, and clinical outcomes of cardiac disease. In this review we synthesize the current understanding of endothelial function and dysfunction as mediators of the cardiomyocyte phenotype in the setting of distinct cardiac pathologies; outline existing in vivo and in vitro models where key features of endothelial cell dysfunction can be recapitulated; and discuss future directions for development of endothelium-targeted therapeutics for cardiac diseases with limited existing treatment options.
Collapse
Affiliation(s)
- Jules D. Allbritton-King
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - Guillermo García-Cardeña
- Department of Pathology, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA, United States
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| |
Collapse
|
8
|
Cortes MB, da Silva RSN, de Oliveira PC, da Silva DS, Irigoyen MCC, Waclawovsky G, Schaun MI. Effect of aerobic and resistance exercise training on endothelial function in individuals with overweight and obesity: a systematic review with meta-analysis of randomized clinical trials. Sci Rep 2023; 13:11826. [PMID: 37479727 PMCID: PMC10362025 DOI: 10.1038/s41598-023-38603-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 07/11/2023] [Indexed: 07/23/2023] Open
Abstract
The objective of this systematic review was to examine the effects of exercise training on endothelial function in individuals with overweight and obesity. Our review study included only randomized controlled trials (RCTs) involving adults (≥ 18 years of age) with body mass index (BMI) ≥ 25.0 kg/m2. Our search was conducted in the electronic bases MEDLINE (PubMed), Cochrane, LILACS and EMBASE and in the gray literature. We performed random-effects analyses for effect estimates and used 95% prediction intervals (95% PI) for estimating the uncertainty of the study results. There were selected 10 RCTs involving 14 groups (n = 400). The quality assessment of studies using Cochrane risk-of-bias 2 (RoB 2) tool identified some concerns. Exercise training resulted in improved flow-mediated dilation (FMD) in individuals with overweight and obesity (p < 0.001) compared to the no-exercise control group. This effect of training modalities on FMD was seen for aerobic training (p < 0.001) but not for resistance training (p = 0.051). There was no difference in FMD in response to exercise training by BMI classification (overweight, obesity, overweight + obesity), p = 0.793. The present results are consistent with the notion that aerobic exercise training elicits favorable adaptations in endothelial function in individuals with overweight and obesity. Our findings should be interpreted with caution because of the small number of studies included in this review.
Collapse
Affiliation(s)
- Maiquel Bueno Cortes
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Porto Alegre, Brasil
| | | | | | - Diego Silveira da Silva
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Porto Alegre, Brasil
| | - Maria Claudia Costa Irigoyen
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Porto Alegre, Brasil
- Experimental Laboratory of Hypertension, Instituto do Coração (InCor), University of São Paulo (USP), São Paulo, Brazil
| | - Gustavo Waclawovsky
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Porto Alegre, Brasil
| | - Maximiliano Isoppo Schaun
- Instituto de Cardiologia do Rio Grande do Sul/Fundação Universitária de Cardiologia, Porto Alegre, Brasil.
- Universidade Luterana do Brasil, Rio Grande do Sul, Brazil.
| |
Collapse
|
9
|
Menon SN, Zerin F, Ezewudo E, Simon NP, Menon SN, Daniel ML, Green AJ, Pandey A, Mackay CE, Hafez S, Moniri NH, Hasan R. Neflamapimod inhibits endothelial cell activation, adhesion molecule expression, leukocyte attachment and vascular inflammation by inhibiting p38 MAPKα and NF-κB signaling. Biochem Pharmacol 2023:115683. [PMID: 37429422 DOI: 10.1016/j.bcp.2023.115683] [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: 04/12/2023] [Revised: 07/06/2023] [Accepted: 07/06/2023] [Indexed: 07/12/2023]
Abstract
Neflamapimod, a selective inhibitor of the alpha isoform of p38 mitogen-activated protein kinase (MAPKα), was investigated for its potential to inhibit lipopolysaccharide (LPS)-induced activation of endothelial cells (ECs), adhesion molecule induction, and subsequent leukocyte attachment to EC monolayers. These events are known to contribute to vascular inflammation and cardiovascular dysfunction. Our results demonstrate that LPS treatment of cultured ECs and rats leads to significant upregulation of adhesion molecules, both in vitro and in vivo, which can be effectively inhibited by Neflamapimod treatment. Western blotting data further reveals that Neflamapimod inhibits LPS-induced phosphorylation of p38 MAPKα and the activation of NF-κB signaling in ECs. Additionally, leukocyte adhesion assays demonstrate a substantial reduction in leukocyte attachment to cultured ECs and the aorta lumen of rats treated with Neflamapimod. Consistent with vascular inflammation, LPS-treated rat arteries exhibit significantly diminished vasodilation response to acetylcholine, however, arteries from rats treated with Neflamapimod maintain their vasodilation capacity, demonstrating its ability to limit LPS-induced vascular inflammation. Overall, our data demonstrate that Neflamapimod effectively inhibits endothelium activation, adhesion molecule expression, and leukocyte attachment, thereby reducing vascular inflammation.
Collapse
Affiliation(s)
- Sreelakshmi N Menon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Farzana Zerin
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Emmanuella Ezewudo
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Nimi P Simon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Sreeranjini N Menon
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Morgan L Daniel
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Andrea J Green
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Ajay Pandey
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; Department of Biological Sciences, Augusta University, Augusta, GA, USA
| | | | - Sherif Hafez
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; Department of Biological Sciences, Augusta University, Augusta, GA, USA
| | - Raquibul Hasan
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA; Department of Biomedical Sciences, School of Medicine, Mercer University, Macon, GA, USA.
| |
Collapse
|
10
|
Murad HA, Alqurashi TM. A novel nano-formulation of olmesartan medoxomil with improved delivery and efficacy in the treatment of indomethacin-induced duodenitis in rats. Braz J Med Biol Res 2023; 56:e12665. [PMID: 37255094 DOI: 10.1590/1414-431x2023e12665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 04/22/2023] [Indexed: 06/01/2023] Open
Abstract
There are few studies addressing duodenal inflammation. This study was designed to investigate the effects of a recently developed biotechnological product, a nano-formulation of olmesartan medoxomil (OM) - olmesartan medoxomil zeinmersomes (OMZ) - for the treatment of indomethacin-induced duodenitis in rats. Adult male Wistar rats were given indomethacin (10 mg/kg/day) for four weeks. They were divided into a positive control group (PC, untreated) and two groups treated orally with 3 mg/kg per day of OM or OMZ for the last two weeks of the 4-week indomethacin-treatment. At end of the four weeks, blood and duodenum were collected. Duodenal homogenate was used for measurement of levels of myeloperoxidase, tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), malondialdehyde, reduced glutathione (GSH), and cleaved caspase-3. Duodenal sections were stained with H&E. Gene expressions of nuclear factor kappa B (NF-κB p65), Bcl-2-associated X protein (Bax), and B-cell lymphoma 2 (Bcl-2) by RT-PCR, and protein expression of survivin by western blot were assessed. Plasma and duodenal olmesartan concentrations were measured by high performance liquid chromatography mass spectrometry. The duodenitis rats showed significantly higher duodenal levels of myeloperoxidase, TNF-α, IL-6, malondialdehyde, and cleaved caspase-3, a significantly lower GSH level, and histopathological alterations. Moreover, they showed upregulated gene expressions of NF-κB p65 and Bax, downregulated gene expression of Bcl-2, decreased Bcl-2/Bax ratio, and lower protein expression of survivin. OMZ was more effective in protecting the duodenum from indomethacin-induced injuries compared to OM due to improved delivery, higher bioavailability, and better anti-inflammatory, antioxidant, and antiapoptotic effects. OMZ could be a better choice for hypertensive patients with non-steroidal anti-inflammatory drugs-induced duodenitis.
Collapse
Affiliation(s)
- H A Murad
- Department of Pharmacology, Faculty of Medicine, Rabigh campus, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - T M Alqurashi
- Department of Pharmacology, Faculty of Medicine, Rabigh campus, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
11
|
Devaux CA, Lagier JC. Unraveling the Underlying Molecular Mechanism of 'Silent Hypoxia' in COVID-19 Patients Suggests a Central Role for Angiotensin II Modulation of the AT1R-Hypoxia-Inducible Factor Signaling Pathway. J Clin Med 2023; 12:jcm12062445. [PMID: 36983445 PMCID: PMC10056466 DOI: 10.3390/jcm12062445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
A few days after being infected with SARS-CoV-2, a fraction of people remain asymptomatic but suffer from a decrease in arterial oxygen saturation in the absence of apparent dyspnea. In light of our clinical investigation on the modulation of molecules belonging to the renin angiotensin system (RAS) in COVID-19 patients, we propose a model that explains 'silent hypoxia'. The RAS imbalance caused by SARS-CoV-2 results in an accumulation of angiotensin 2 (Ang II), which activates the angiotensin 2 type 1 receptor (AT1R) and triggers a harmful cascade of intracellular signals leading to the nuclear translocation of the hypoxia-inducible factor (HIF)-1α. HIF-1α transactivates many genes including the angiotensin-converting enzyme 1 (ACE1), while at the same time, ACE2 is downregulated. A growing number of cells is maintained in a hypoxic condition that is self-sustained by the presence of the virus and the ACE1/ACE2 ratio imbalance. This is associated with a progressive worsening of the patient's biological parameters including decreased oxygen saturation, without further clinical manifestations. When too many cells activate the Ang II-AT1R-HIF-1α axis, there is a 'hypoxic spillover', which marks the tipping point between 'silent' and symptomatic hypoxia in the patient. Immediate ventilation is required to prevent the 'hypoxic spillover'.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
- Centre National de la Recherche Scientifique, 13000 Marseille, France
| | - Jean-Christophe Lagier
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
| |
Collapse
|
12
|
Margaroli C, Benson P, Gastanadui MG, Song C, Viera L, Xing D, Wells JM, Patel R, Gaggar A, Payne GA. Spatial transcriptomic profiling of coronary endothelial cells in SARS-CoV-2 myocarditis. Front Med (Lausanne) 2023; 10:1118024. [PMID: 36968839 PMCID: PMC10034160 DOI: 10.3389/fmed.2023.1118024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 02/13/2023] [Indexed: 03/29/2023] Open
Abstract
Objectives Our objective was to examine coronary endothelial and myocardial programming in patients with severe COVID-19 utilizing digital spatial transcriptomics. Background Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has well-established links to thrombotic and cardiovascular events. Endothelial cell infection was initially proposed to initiate vascular events; however, this paradigm has sparked growing controversy. The significance of myocardial infection also remains unclear. Methods Autopsy-derived cardiac tissue from control (n = 4) and COVID-19 (n = 8) patients underwent spatial transcriptomic profiling to assess differential expression patterns in myocardial and coronary vascular tissue. Our approach enabled transcriptional profiling in situ with preserved anatomy and unaltered local SARS-CoV-2 expression. In so doing, we examined the paracrine effect of SARS-CoV-2 infection in cardiac tissue. Results We observed heterogeneous myocardial infection that tended to colocalize with CD31 positive cells within coronary capillaries. Despite these differences, COVID-19 patients displayed a uniform and unique myocardial transcriptional profile independent of local viral burden. Segmentation of tissues directly infected with SARS-CoV-2 showed unique, pro-inflammatory expression profiles including upregulated mediators of viral antigen presentation and immune regulation. Infected cell types appeared to primarily be capillary endothelial cells as differentially expressed genes included endothelial cell markers. However, there was limited differential expression within the endothelium of larger coronary vessels. Conclusion Our results highlight altered myocardial programming during severe COVID-19 that may in part be associated with capillary endothelial cells. However, similar patterns were not observed in larger vessels, diminishing endotheliitis, and endothelial activation as key drivers of cardiovascular events during COVID-19.
Collapse
Affiliation(s)
- Camilla Margaroli
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Paul Benson
- Department of Pathology, Division of Anatomic Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Maria G. Gastanadui
- Cardiopulmonary Research Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Chunyan Song
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Cardiopulmonary Research Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Liliana Viera
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Dongqi Xing
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Cardiopulmonary Research Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, United States
| | - J. Michael Wells
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Cardiopulmonary Research Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Medical Service at Birmingham VA Medical Center, Birmingham, AL, United States
| | - Rakesh Patel
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Pathology, Division of Molecular and Cellular Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Amit Gaggar
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Cardiopulmonary Research Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Lung Health Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Medical Service at Birmingham VA Medical Center, Birmingham, AL, United States
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Gregory A. Payne
- Program in Protease/Matrix Biology, University of Alabama at Birmingham, Birmingham, AL, United States
- Cardiopulmonary Research Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL, United States
- Vascular Biology and Hypertension Program, University of Alabama at Birmingham, Birmingham, AL, United States
- Medical Service at Birmingham VA Medical Center, Birmingham, AL, United States
- Comprehensive Cardiovascular Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
13
|
Arnhold J. Host-Derived Cytotoxic Agents in Chronic Inflammation and Disease Progression. Int J Mol Sci 2023; 24:ijms24033016. [PMID: 36769331 PMCID: PMC9918110 DOI: 10.3390/ijms24033016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
At inflammatory sites, cytotoxic agents are released and generated from invading immune cells and damaged tissue cells. The further fate of the inflammation highly depends on the presence of antagonizing principles that are able to inactivate these host-derived cytotoxic agents. As long as the affected tissues are well equipped with ready-to-use protective mechanisms, no damage by cytotoxic agents occurs and resolution of inflammation is initiated. However, long-lasting and severe immune responses can be associated with the decline, exhaustion, or inactivation of selected antagonizing principles. Hence, cytotoxic agents are only partially inactivated and contribute to damage of yet-unperturbed cells. Consequently, a chronic inflammatory process results. In this vicious circle of permanent cell destruction, not only novel cytotoxic elements but also novel alarmins and antigens are liberated from affected cells. In severe cases, very low protection leads to organ failure, sepsis, and septic shock. In this review, the major classes of host-derived cytotoxic agents (reactive species, oxidized heme proteins and free heme, transition metal ions, serine proteases, matrix metalloproteases, and pro-inflammatory peptides), their corresponding protective principles, and resulting implications on the pathogenesis of diseases are highlighted.
Collapse
Affiliation(s)
- Jürgen Arnhold
- Medical Faculty, Institute of Medical Physics and Biophysics, Leipzig University, Härtelstr. 16-18, 04107 Leipzig, Germany
| |
Collapse
|
14
|
Dong CX, Malecki C, Robertson E, Hambly B, Jeremy R. Molecular Mechanisms in Genetic Aortopathy-Signaling Pathways and Potential Interventions. Int J Mol Sci 2023; 24:ijms24021795. [PMID: 36675309 PMCID: PMC9865322 DOI: 10.3390/ijms24021795] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/02/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Thoracic aortic disease affects people of all ages and the majority of those aged <60 years have an underlying genetic cause. There is presently no effective medical therapy for thoracic aneurysm and surgery remains the principal intervention. Unlike abdominal aortic aneurysm, for which the inflammatory/atherosclerotic pathogenesis is well established, the mechanism of thoracic aneurysm is less understood. This paper examines the key cell signaling systems responsible for the growth and development of the aorta, homeostasis of endothelial and vascular smooth muscle cells and interactions between pathways. The evidence supporting a role for individual signaling pathways in pathogenesis of thoracic aortic aneurysm is examined and potential novel therapeutic approaches are reviewed. Several key signaling pathways, notably TGF-β, WNT, NOTCH, PI3K/AKT and ANGII contribute to growth, proliferation, cell phenotype and survival for both vascular smooth muscle and endothelial cells. There is crosstalk between pathways, and between vascular smooth muscle and endothelial cells, with both synergistic and antagonistic interactions. A common feature of the activation of each is response to injury or abnormal cell stress. Considerable experimental evidence supports a contribution of each of these pathways to aneurysm formation. Although human information is less, there is sufficient data to implicate each pathway in the pathogenesis of human thoracic aneurysm. As some pathways i.e., WNT and NOTCH, play key roles in tissue growth and organogenesis in early life, it is possible that dysregulation of these pathways results in an abnormal aortic architecture even in infancy, thereby setting the stage for aneurysm development in later life. Given the fine tuning of these signaling systems, functional polymorphisms in key signaling elements may set up a future risk of thoracic aneurysm. Multiple novel therapeutic agents have been developed, targeting cell signaling pathways, predominantly in cancer medicine. Future investigations addressing cell specific targeting, reduced toxicity and also less intense treatment effects may hold promise for effective new medical treatments of thoracic aortic aneurysm.
Collapse
Affiliation(s)
- Charlotte Xue Dong
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Cassandra Malecki
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
| | - Elizabeth Robertson
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Brett Hambly
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
| | - Richmond Jeremy
- Faculty of Health and Medical Sciences, University of Sydney, Sydney, NSW 2006, Australia
- The Baird Institute, Camperdown, NSW 2042, Australia
- Correspondence:
| |
Collapse
|
15
|
Bravo FI, Calvo E, López-Villalba RA, Torres-Fuentes C, Muguerza B, García-Ruiz A, Morales D. Valorization of Chicken Slaughterhouse Byproducts to Obtain Antihypertensive Peptides. Nutrients 2023; 15:457. [PMID: 36678328 PMCID: PMC9864718 DOI: 10.3390/nu15020457] [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: 12/22/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Hypertension (HTN) is the leading cause of premature deaths worldwide and the main preventable risk factor for cardiovascular diseases. Therefore, there is a current need for new therapeutics to manage this condition. In this regard, protein hydrolysates containing antihypertensive bioactive peptides are of increasing interest. Thus, agri-food industry byproducts have emerged as a valuable source to obtain these hydrolysates as they are rich in proteins and inexpensive. Among these, byproducts from animal origin stand out as they are abundantly generated worldwide. Hence, this review is focused on evaluating the potential role of chicken slaughterhouse byproducts as a source of peptides for managing HTN. Several of these byproducts such as blood, bones, skins, and especially, chicken feet have been used to obtain protein hydrolysates with angiotensin-converting enzyme (ACE)-inhibitory activity and blood pressure-lowering effects. An increase in levels of endogenous antioxidant compounds, a reduction in ACE activity, and an improvement of HTN-associated endothelial dysfunction were the mechanisms underlying their effects. However, most of these studies were carried out in animal models, and further clinical studies are needed in order to confirm these antihypertensive properties. This would increase the value of these byproducts, contributing to the circular economy model of slaughterhouses.
Collapse
Affiliation(s)
| | | | | | | | | | - Almudena García-Ruiz
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | | |
Collapse
|
16
|
Tissue Derivation and Biological Sex Uniquely Mediate Endothelial Cell Protein Expression, Redox Status, and Nitric Oxide Synthesis. Cells 2022; 12:cells12010093. [PMID: 36611888 PMCID: PMC9818567 DOI: 10.3390/cells12010093] [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: 12/08/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Human endothelial cells are routinely utilized in cardiovascular research to provide a translational foundation for understanding how the vascular endothelium functions in vivo. However, little attention has been given to whether there are sex specific responses in vitro. Similarly, it is unclear whether endothelial cells derived from distinct tissues behave in a homogenous manner. Herein, we demonstrate that marked sex differences exist within, and between, commonly utilized human primary endothelial cells from healthy donors, with respect to redox status, nitric oxide synthesis, and associated proteins that can mediate their expression. Further, we demonstrate that endothelial cells respond uniquely to inflammatory insult in a sex- and tissue origin-dependent manner. Our findings suggest sex and tissue derivation may need to be considered when studying endothelial cells in vitro as cells derived from distinct tissue and sexes may not behave interchangeably.
Collapse
|
17
|
Abstract
Purpose: COVID-19, a novel infection, presented with several complications, including socioeconomical and reproductive health challenges such as erectile dysfunction (ED). The present review summarizes the available shreds of evidence on the impact of COVID-19 on ED.Materials and methods: All published peer-reviewed articles from the onset of the COVID-19 outbreak to date, relating to ED, were reviewed. Results: Available pieces of evidence that ED is a consequence of COVID-19 are convincing. COVID-19 and ED share common risk factors such as disruption of vascular integrity, cardiovascular disease (CVD), cytokine storm, diabetes, obesity, and chronic kidney disease (CKD). COVID-19 also induces impaired pulmonary haemodynamics, increased ang II, testicular damage and low serum testosterone, and reduced arginine-dependent NO bioavailability that promotes reactive oxygen species (ROS) generation and endothelial dysfunction, resulting in ED. In addition, COVID-19 triggers psychological/mental stress and suppresses testosterone-dependent dopamine concentration, which contributes to incident ED.Conclusions: In conclusion, COVID-19 exerts a detrimental effect on male reproductive function, including erectile function. This involves a cascade of events from multiple pathways. As the pandemic dwindles, identifying the long-term effects of COVID-19-induced ED, and proffering adequate and effective measures in militating against COVID-19-induced ED remains pertinent.
Collapse
Affiliation(s)
- D H Adeyemi
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Osun State University, Nigeria
| | - A F Odetayo
- Department of Physiology, University of Ilorin, Ilorin, Nigeria
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
| | - M A Hamed
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- The Brainwill Laboratories, Osogbo, Nigeria
- Department of Medical Laboratory Sciences, College of Medicine and Health Sciences, Afe Babalola University, Ado-Ekiti, Nigeria
| | - R E Akhigbe
- Reproductive Biology and Toxicology Research Laboratory, Oasis of Grace Hospital, Osogbo, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
18
|
Devaux CA, Camoin-Jau L. An update on angiotensin-converting enzyme 2 structure/functions, polymorphism, and duplicitous nature in the pathophysiology of coronavirus disease 2019: Implications for vascular and coagulation disease associated with severe acute respiratory syndrome coronavirus infection. Front Microbiol 2022; 13:1042200. [PMID: 36519165 PMCID: PMC9742611 DOI: 10.3389/fmicb.2022.1042200] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 11/07/2022] [Indexed: 08/01/2023] Open
Abstract
It has been known for many years that the angiotensin-converting enzyme 2 (ACE2) is a cell surface enzyme involved in the regulation of blood pressure. More recently, it was proven that the severe acute respiratory syndrome coronavirus (SARS-CoV-2) interacts with ACE2 to enter susceptible human cells. This functional duality of ACE2 tends to explain why this molecule plays such an important role in the clinical manifestations of coronavirus disease 2019 (COVID-19). At the very start of the pandemic, a publication from our Institute (entitled "ACE2 receptor polymorphism: susceptibility to SARS-CoV-2, hypertension, multi-organ failure, and COVID-19 disease outcome"), was one of the first reviews linking COVID-19 to the duplicitous nature of ACE2. However, even given that COVID-19 pathophysiology may be driven by an imbalance in the renin-angiotensin system (RAS), we were still far from understanding the complexity of the mechanisms which are controlled by ACE2 in different cell types. To gain insight into the physiopathology of SARS-CoV-2 infection, it is essential to consider the polymorphism and expression levels of the ACE2 gene (including its alternative isoforms). Over the past 2 years, an impressive amount of new results have come to shed light on the role of ACE2 in the pathophysiology of COVID-19, requiring us to update our analysis. Genetic linkage studies have been reported that highlight a relationship between ACE2 genetic variants and the risk of developing hypertension. Currently, many research efforts are being undertaken to understand the links between ACE2 polymorphism and the severity of COVID-19. In this review, we update the state of knowledge on the polymorphism of ACE2 and its consequences on the susceptibility of individuals to SARS-CoV-2. We also discuss the link between the increase of angiotensin II levels among SARS-CoV-2-infected patients and the development of a cytokine storm associated microvascular injury and obstructive thrombo-inflammatory syndrome, which represent the primary causes of severe forms of COVID-19 and lethality. Finally, we summarize the therapeutic strategies aimed at preventing the severe forms of COVID-19 that target ACE2. Changing paradigms may help improve patients' therapy.
Collapse
Affiliation(s)
- Christian A. Devaux
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Center National de la Recherche Scientifique, Marseille, France
| | - Laurence Camoin-Jau
- Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Laboratoire d’Hématologie, Hôpital de La Timone, APHM, Boulevard Jean-Moulin, Marseille, France
| |
Collapse
|
19
|
Margaroli C, Benson P, Gastanadui MG, Song C, Viera L, Xing D, Wells JM, Patel R, Gaggar A, Payne GA. Spatial transcriptomic profiling of coronary endothelial cells in SARS-CoV-2 myocarditis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.09.25.509426. [PMID: 36203548 PMCID: PMC9536040 DOI: 10.1101/2022.09.25.509426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
OBJECTIVES Our objective was to examine coronary endothelial and myocardial programming in patients with severe COVID-19 utilizing digital spatial transcriptomics. BACKGROUND Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has well-established links to thrombotic and cardiovascular events. Endothelial cell infection was initially proposed to initiate vascular events; however, this paradigm has sparked growing controversy. The significance of myocardial infection also remains unclear. METHODS Autopsy-derived cardiac tissue from control (n = 4) and COVID-19 (n = 8) patients underwent spatial transcriptomic profiling to assess differential expression patterns in myocardial and coronary vascular tissue. Our approach enabled transcriptional profiling in situ with preserved anatomy and unaltered local SARS-CoV-2 expression. In so doing, we examined the paracrine effect of SARS-CoV-2 infection in cardiac tissue. RESULTS We observed heterogeneous myocardial infection that tended to colocalize with CD31 positive cells within coronary capillaries. Despite these differences, COVID-19 patients displayed a uniform and unique myocardial transcriptional profile independent of local viral burden. Segmentation of tissues directly infected with SARS-CoV-2 showed unique, pro-inflammatory expression profiles including upregulated mediators of viral antigen presentation and immune regulation. Infected cell types appeared to primarily be capillary endothelial cells as differentially expressed genes included endothelial cell markers. However, there was limited differential expression within the endothelium of larger coronary vessels. CONCLUSIONS Our results highlight altered myocardial programming during severe COVID-19 that may in part be associated with capillary endothelial cells. However, similar patterns were not observed in larger vessels, diminishing endotheliitis and endothelial activation as key drivers of cardiovascular events during COVID-19. CONDENSED ABSTRACT SARS-CoV-2 is linked to thrombotic and cardiovascular events; however, the mechanism remains uncertain. Our objective was to examine coronary endothelial and myocardial programming in patients with severe COVID-19 utilizing digital spatial transcriptomics. Autopsy-derived coronary arterial and cardiac tissues from control and COVID-19 patients underwent spatial transcriptomic profiling. Our approach enabled transcriptional profiling in situ with preserved anatomy and unaltered local SARS-CoV-2 expression. We observed unique, pro-inflammatory expression profiles among all COVID-19 patients. While heterogeneous viral expression was noted within the tissue, SARS-CoV-2 tended to colocalize with CD31 positive cells within coronary capillaries and was associated with unique expression profiles. Similar patterns were not observed in larger coronary vessels. Our results highlight altered myocardial programming during severe COVID-19 that may in part be associated with capillary endothelial cells. Such results diminish coronary arterial endotheliitis and endothelial activation as key drivers of cardiovascular events during COVID-19 infection. LIST OF HIGHLIGHTS SARS-CoV-2 has variable expression patterns within the myocardium of COVID-19 patientsSARS-CoV-2 infection induces a unique myocardial transcriptional programming independent of local viral burdenSARS-CoV-2 myocarditis is predominantly associated with capillaritis, and tissues directly infected with SARS-CoV-2 have unique, pro-inflammatory expression profilesDiffuse endothelial activation of larger coronary vessels was absent, diminishing large artery endotheliitis as a significant contributor to cardiovascular events during COVID-19 infection.
Collapse
|
20
|
Yi J, Miao J, Zuo Q, Owusu F, Dong Q, Lin P, Wang Q, Gao R, Kong X, Yang L. COVID-19 pandemic: A multidisciplinary perspective on the pathogenesis of a novel coronavirus from infection, immunity and pathological responses. Front Immunol 2022; 13:978619. [PMID: 36091053 PMCID: PMC9459044 DOI: 10.3389/fimmu.2022.978619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/04/2022] [Indexed: 12/15/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus2 (SARS-CoV-2), has spread to more than 200 countries and regions, having a huge impact on human health, hygiene, and economic activities. The epidemiological and clinical phenotypes of COVID-19 have increased since the onset of the epidemic era, and studies into its pathogenic mechanisms have played an essential role in clinical treatment, drug development, and prognosis prevention. This paper reviews the research progress on the pathogenesis of the novel coronavirus (SARS-CoV-2), focusing on the pathogenic characteristics, loci of action, and pathogenic mechanisms leading to immune response malfunction of SARS-CoV-2, as well as summarizing the pathological damage and pathological manifestations it causes. This will update researchers on the latest SARS-CoV-2 research and provide directions for future therapeutic drug development.
Collapse
Affiliation(s)
- Jia Yi
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jiameng Miao
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingwei Zuo
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Felix Owusu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qiutong Dong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Peizhe Lin
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Gao
- Institute of Clinical Pharmacology of Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xianbin Kong
- College of Traditional Chinese medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Long Yang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
21
|
Transcriptomics of angiotensin II-induced long noncoding and coding RNAs in endothelial cells. J Hypertens 2022; 40:1303-1313. [PMID: 35762471 DOI: 10.1097/hjh.0000000000003140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Angiotensin II (Ang II)-induced endothelial dysfunction plays an important role in the pathogenesis of cardiovascular diseases such as systemic hypertension, cardiac hypertrophy and atherosclerosis. Recently, long noncoding RNAs (lncRNAs) have been shown to play an essential role in the pathobiology of cardiovascular diseases; however, the effect of Ang II on lncRNAs and coding RNAs expression in endothelial cells has not been evaluated. Accordingly, we sought to evaluate the expression profiles of lncRNAs and coding RNAs in endothelial cells following treatment with Ang II. METHODS Human umbilical vein endothelial cells (HUVECs) were cultured and treated with Ang II (10-6 mol/l) for 24 h. The cells were then profiled for the expression of lncRNAs and mRNAs using the Arraystar Human lncRNA Expression Microarray V3.0. RESULTS In HUVECs following Ang II treatment, from a total of 30 584 lncRNA targets screened, 25 targets were significantly upregulated, while 69 were downregulated. In the same HUVECs samples, from 26 106 mRNA targets screened, 28 targets were significantly upregulated and 67 were downregulated. Of the differentially expressed lncRNAs, RP11-354P11.2 and RP11-360F5.1 were the most upregulated (11-fold) and downregulated (three-fold) lncRNAs, respectively. Assigning the differentially regulated genes into functional groups using bioinformatics reveals numerous genes involved in the nucleotide excision repair and ECM-receptor interaction. CONCLUSION This is the first study to profile the Ang II-induced differentially expressed lncRNAs and mRNAs in human endothelial cells. Our results reveal novel targets and substantially extend the list of potential candidate genes involved in Ang II-induced endothelial dysfunction and cardiovascular diseases.
Collapse
|
22
|
Bioactive Natural Products against Systemic Arterial Hypertension: A Past 20-Year Systematic and Prospective Review. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:8499625. [PMID: 35769156 PMCID: PMC9236778 DOI: 10.1155/2022/8499625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/24/2022] [Indexed: 11/18/2022]
Abstract
Background. Systemic arterial hypertension is one of the most common cardiovascular risks, corresponding to 45% of deaths involving CVDs. The use of natural products, such as medicinal plants, belongs to a millennial part of human therapeutics history and has been employed as an alternative anti-hypertensive treatment. Objective. The present review aims to prospect some natural products already experimentally assayed against arterial hypertension through scientific virtual libraries and patent documents over the past 20 years. Search strategy. This is a systematic review of the adoption of the PRISMA protocol and a survey of the scientific literature that synthesizes the results from published articles between 2001 and 2020 concerning the use of medicinal plants in the management of hypertension, including which parts of the plant or organism are used, as well as the mechanisms of action underlying the anti-hypertensive effect. Furthermore, a technological prospection was also carried out in patent offices from different countries in order to check technologies based on natural products claimed for the treatment or prevention of hypertension. Inclusion criteria. Scientific articles where a natural product had been experimentally assayed for anti-hypertensive activity (part of plants, plant extracts, and products derived from other organisms) were included. Data extraction and analysis. The selected abstracts of the articles and patent documents were submitted to a rigorous reading process. Those articles and patents that were not related to anti-hypertensive effects and claimed potential applications were excluded from the search. Results. Eighty specimens of biological species that showed anti-hypertensive activity were recovered, with 01 representative from the kingdom Fungi and 02 from the kingdom Protista, with emphasis on the families Asteraceae and Lamiaceae, with 6 representatives each. Leaves and aerial parts were the most used parts of the plants for the extraction of anti-hypertensive products, with maceration being the most used extraction method. Regarding phytochemical analyses, the most described classes of biomolecules in the reviewed works were alkaloids, terpenes, coumarins, flavonoids, and peptides, with the reduction of oxidative stress and the release of NO among the mechanisms of action most involved in this process. Regarding the number of patent filings, China was the country that stood out as the main one, with 813 registrations. Conclusion. The anti-hypertensive activity of natural products is still little explored in Western countries. Besides, China and India have shown more results in this area than other countries, confirming the strong influence of traditional medicine in these countries.
Collapse
|
23
|
Ibarz-Blanch N, Morales D, Calvo E, Ros-Medina L, Muguerza B, Bravo FI, Suárez M. Role of Chrononutrition in the Antihypertensive Effects of Natural Bioactive Compounds. Nutrients 2022; 14:nu14091920. [PMID: 35565887 PMCID: PMC9103085 DOI: 10.3390/nu14091920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 04/26/2022] [Accepted: 05/02/2022] [Indexed: 12/13/2022] Open
Abstract
Hypertension (HTN) is one of the main cardiovascular risk factors and is considered a major public health problem. Numerous approaches have been developed to lower blood pressure (BP) in hypertensive patients, most of them involving pharmacological treatments. Within this context, natural bioactive compounds have emerged as a promising alternative to drugs in HTN prevention. This work reviews not only the mechanisms of BP regulation by these antihypertensive compounds, but also their efficacy depending on consumption time. Although a plethora of studies has investigated food-derived compounds, such as phenolic compounds or peptides and their impact on BP, only a few addressed the relevance of time consumption. However, it is known that BP and its main regulatory mechanisms show a 24-h oscillation. Moreover, evidence shows that phenolic compounds can interact with clock genes, which regulate the biological rhythm followed by many physiological processes. Therefore, further research might be carried out to completely elucidate the interactions along the time–nutrition–hypertension axis within the framework of chrononutrition.
Collapse
Affiliation(s)
| | | | - Enrique Calvo
- Correspondence: (E.C.); (F.I.B.); Tel.: +34-977558837 (E.C.)
| | | | | | | | | |
Collapse
|
24
|
Wang R, Guo Y, Li L, Luo M, Peng L, Lv D, Cheng Z, Xue Q, Wang L, Huang J. Role of thioredoxin-interacting protein in mediating endothelial dysfunction in hypertension. Genes Dis 2022; 9:753-765. [PMID: 35782967 PMCID: PMC9243351 DOI: 10.1016/j.gendis.2020.08.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/23/2020] [Accepted: 08/23/2020] [Indexed: 11/17/2022] Open
Abstract
Excessive oxidative stress is a major causative factor of endothelial dysfunction in hypertension. As an endogenous pro-oxidant, thioredoxin-interacting protein (TXNIP) contributes to oxidative damage in various tissues. The present study aimed to investigate the role of TXNIP in mediating endothelial dysfunction in hypertension. In vivo, an experimental model of acquired hypertension was established with two-kidney, one-clip (2K1C) surgery. The expression of TXNIP in the vascular endothelial cells of multiple vessels was significantly increased in hypertensive rats compared with sham-operated rats. Resveratrol, a TXNIP inhibitor, suppressed vascular oxidative damage and increased the expression and activity of eNOS in the aorta of hypertensive rats. Notably, impaired endothelium-dependent vasodilation was effectively improved by TXNIP inhibition in hypertensive rats. In vitro, we observed that Ang II increased the expression of TXNIP in primary human aortic endothelial cells (HAECs) and that TXNIP knockdown by RNA interference alleviated cellular oxidative stress damage and mitigated the impaired eNOS activation and intracellular nitric oxide (NO) production observed in Ang II-treated HAECs. However, inhibiting thioredoxin (TRX) with PX-12 completely blunted the protective effect of silencing TXNIP. In addition, TXNIP knockdown facilitated TRX expression and promoted TRX nuclear translocation to further activate AP1 and REF1. TRX overexpression exhibited favorable effects on eNOS/NO homeostasis in Ang II-treated HAECs. Thus, TXNIP contributes to oxidative stress and endothelial dysfunction in hypertension, and these effects are dependent on the antioxidant capacity of TRX, suggesting that targeting TXNIP may be a novel strategy for antihypertensive therapy.
Collapse
Affiliation(s)
- Ruiyu Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Yongzheng Guo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Lingjiao Li
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Linqian Peng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Dingyi Lv
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Zhe Cheng
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, PR China
- Institute of Life Science, Chongqing Medical University, Chongqing 400016, PR China
| | - Qian Xue
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Liang Wang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
| | - Jing Huang
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, PR China
- Corresponding author. Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, No.76 Linjiang Road, Chongqing 400010, PR China. Fax: +86 23 63711527.
| |
Collapse
|
25
|
Borghi C, Levy BI. Synergistic actions between angiotensin-converting enzyme inhibitors and statins in atherosclerosis. Nutr Metab Cardiovasc Dis 2022; 32:815-826. [PMID: 35082055 DOI: 10.1016/j.numecd.2021.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/31/2021] [Accepted: 11/25/2021] [Indexed: 01/09/2023]
Abstract
AIMS Hypertension and hypercholesterolemia are independent risk factors for atherosclerotic cardiovascular disease (ASCVD) by acting directly on the endothelium and activating the renin-angiotensin aldosterone system (RAAS) and mevalonate pathways. This review examines how the severity and duration of these risk factors may influence the cardiovascular risk through a reciprocal interplay leading to oxidative stress and pro-inflammatory response. DATA SYNTHESIS The review highlights the clinical evidence supporting the benefits of statins and angiotensin-converting enzyme (ACE) inhibitors for hypertension, lipid disorders and ASCVD management, both individually and combined, at all stages of the cardiovascular continuum. CONCLUSION Drug strategies incorporating an ACE-inhibitor and a statin, and in particular perindopril and atorvastatin, have consistently demonstrated reductions in the rate of ASCVD events in patients with hypertension and lipid disorders, cementing their position as first-line therapies for the management of atherosclerosis complications.
Collapse
Affiliation(s)
- Claudio Borghi
- Department of Medical and Surgical Sciences, IRCCS-S.Orsola, University of Bologna, Italy.
| | - Bernard I Levy
- INSERM Unit 970, PARCC, 56 rue Leblanc, 75015 Paris, France
| |
Collapse
|
26
|
Ayala-Ramírez P, González M, Escudero C, Quintero-Arciniegas L, Giachini FR, Alves de Freitas R, Damiano AE, García-Robles R. Severe Acute Respiratory Syndrome Coronavirus 2 Infection in Pregnancy. A Non-systematic Review of Clinical Presentation, Potential Effects of Physiological Adaptations in Pregnancy, and Placental Vascular Alterations. Front Physiol 2022; 13:785274. [PMID: 35431989 PMCID: PMC9005899 DOI: 10.3389/fphys.2022.785274] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
In December 2019, the novel Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) rapidly spread to become a pandemic. To date, increasing evidence has described the potential negative impact of SARS-CoV-2 infection on pregnant women. Although the pathophysiology of coronavirus disease 2019 (COVID-19) is not entirely understood, there is emerging evidence that it causes a severe systemic inflammatory response associated with vascular alterations that could be of special interest considering some physiological changes in pregnancy. Additionally, these alterations may affect the physiology of the placenta and are associated with pregnancy complications and abnormal histologic findings. On the other hand, data about the vaccine against SARS-CoV-2 are limited, but the risks of administering COVID-19 vaccines during pregnancy appear to be minimal. This review summarizes the current literature on SARSCoV2 virus infection, the development of COVID-19 and its relationship with physiological changes, and angiotensin-converting enzyme 2 (ACE2) function during pregnancy. We have particularly emphasized evidence coming from Latin American countries.
Collapse
Affiliation(s)
- Paola Ayala-Ramírez
- School of Medicine, Human Genetics Institute, Pontificia Universidad Javeriana, Bogotá, Colombia
- *Correspondence: Paola Ayala-Ramírez,
| | - Marcelo González
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
- Laboratorio de Investigación Materno-Fetal (LIMaF), Departamento de Obstetricia y Ginecología, Facultad de Medicina, Universidad de Concepción, Concepción, Chile
- Marcelo González,
| | - Carlos Escudero
- Group of Research and Innovation in Vascular Health (GRIVAS Health), Chillan, Chile
- Laboratory of Vascular Physiology, Department of Basic Sciences, Faculty of Sciences, Universidad del Bio-Bio, Chillan, Chile
| | - Laura Quintero-Arciniegas
- Perinatal Medicine Seedbed, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
- Department of Physiological Sciences, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Fernanda R. Giachini
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, Brazil
- Institute of Biological Sciences, Federal University of Goias, Goiânia, Brazil
| | | | - Alicia E. Damiano
- Laboratorio de Biología de la Reproducción, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO)- CONICET- Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
- Cátedra de Biología Celular y Molecular, Departamento de Ciencias Biológicas, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Reggie García-Robles
- Department of Physiological Sciences, School of Medicine, Pontificia Universidad Javeriana, Bogotá, Colombia
| |
Collapse
|
27
|
COVID-19: cardiovascular manifestations-a review of the cardiac effects. J Geriatr Cardiol 2022; 19:245-250. [PMID: 35464648 PMCID: PMC9002085 DOI: 10.11909/j.issn.1671-5411.2022.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
28
|
Alimoradi N, Sharqi M, Firouzabadi D, Sadeghi MM, Moezzi MI, Firouzabadi N. SNPs of ACE1 (rs4343) and ACE2 (rs2285666) genes are linked to SARS-CoV-2 infection but not with the severity of disease. Virol J 2022; 19:48. [PMID: 35305693 PMCID: PMC8934128 DOI: 10.1186/s12985-022-01782-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/10/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 and the renin-angiotensin system (RAS) are linked by angiotensin-converting enzyme 2 (ACE2), a key enzyme in RAS that has been validated as a SARS-CoV-2 receptor. Functional ACE1/ACE2 gene polymorphisms may lead to the imbalance between ACE/ACE2 ratio and thus generating RAS imbalance that is associated with higher degrees of lung damage in ARDS that may contribute to the COVID-19 infection outcome. Herein, we investigated the role of RAS gene polymorphisms, ACE1 (A2350G) and ACE2 (G8790A) as risk predictors for susceptibility and severity of COVID-19 infection. A total of 129 included: negative controls without a history of COVID-19 infection (n = 50), positive controls with a history of COVID-19 infection who were not hospitalized (n = 35), and patients with severe COVID-19 infection who were hospitalized in the intensive care unit (n = 44). rs4343 of ACE and rs2285666 of ACE2 were genotyped using PCR-RFLP method. Our results indicated that susceptibility to COVID-19 infection was associated with age, GG genotype of A2350G (Pa = 0.01; OR 4.7; 95% CI 1.4-15.1 and Pc = 0.040; OR 2.5; 95% CI 1.05-6.3) and GG genotype of G8790A (Pa = 0.044; OR 6.17; 95% CI 1.05-35.71 and Pc = 0.0001; OR 5.5; 95% CI 2.4-12.4). The G allele of A2350G (Pa = 0.21; OR 1.74; 95% CI 0.73-4.17 and Pc = 0.007; OR 2.1; 95% CI 1.2-3.5) and G allele of G8790A (Pa = 0.002; OR 4.26; 95% CI 1.7-10.65 and Pc = 0.0001; OR 4.7; 95% CI 2.4-9.2) were more frequent in ICU-admitted patients and positive control group. Also lung involvement due to COVID-19 infection was associated with age and the comorbidities such as diabetes. In conclusion, our findings support the association between the wild genotype (GG) of ACE2 and homozygote genotype (GG) of ACE1 and sensitivity to COVID-19 infection, but not its severity. However, confirmation of this hypothesis requires further studies with more participants.
Collapse
Affiliation(s)
- Nahid Alimoradi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Moein Sharqi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Dena Firouzabadi
- Shahid Faghihi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
- Clinical Pharmacy Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Moein Sadeghi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Iman Moezzi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
29
|
Zhu YC, Liang B, Gu N. Cellular and Molecular Mechanism of Traditional Chinese Medicine on Ventricular Remodeling. Front Cardiovasc Med 2021; 8:753095. [PMID: 34926607 PMCID: PMC8671630 DOI: 10.3389/fcvm.2021.753095] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Ventricular remodeling is related to the renin-angiotensin-aldosterone system, immune system, and various cytokines involved in inflammation, apoptosis, and cell signal regulation. Accumulated studies have shown that traditional Chinese medicine can significantly inhibit the process of ventricular remodeling, which may be related to the mechanism mentioned above. Here, we conducted a system overview to critically review the cellular and molecular mechanism of traditional Chinese medicine on ventricular remodeling. We mainly searched PubMed for basic research about the anti-ventricular remodeling of traditional Chinese medicine in 5 recent years, and then objectively summarized these researches. We included more than 25 kinds of Chinese herbal medicines including Qi-Li-Qian-Xin, Qi-Shen-Yi-Qi Pill, Xin-Ji-Er-Kang Formula, and Yi-Qi-Wen-Yang Decoction, and found that they can inhibit ventricular remodeling effectively through multi-components and multi-action targets, which are promoting the clinical application of traditional Chinese medicine.
Collapse
Affiliation(s)
- Yong-Chun Zhu
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Bo Liang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Ning Gu
- Nanjing Hospital of Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
30
|
Cai S, Pan N, Xu M, Su Y, Qiao K, Chen B, Zheng B, Xiao M, Liu Z. ACE Inhibitory Peptide from Skin Collagen Hydrolysate of Takifugu bimaculatus as Potential for Protecting HUVECs Injury. Mar Drugs 2021; 19:md19120655. [PMID: 34940654 PMCID: PMC8703921 DOI: 10.3390/md19120655] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/20/2021] [Accepted: 11/22/2021] [Indexed: 12/21/2022] Open
Abstract
Angiotensin-I-converting enzyme (ACE) is a crucial enzyme or receptor that catalyzes the generation of potent vasopressor angiotensin II (Ang II). ACE inhibitory peptides from fish showed effective ACE inhibitory activity. In this study, we reported an ACE inhibitory peptide from Takifugu bimaculatus (T. bimaculatus), which was obtained by molecular docking with acid-soluble collagen (ASC) hydrolysate of T. bimaculatus. The antihypertensive effects and potential mechanism were conducted using Ang-II-induced human umbilical vein endothelial cells (HUVECs) as a model. The results showed that FNLRMQ alleviated the viability and facilitated apoptosis of Ang-II-induced HUVECs. Further research suggested that FNLRMQ may protect Ang-II-induced endothelial injury by regulating Nrf2/HO-1 and PI3K/Akt/eNOS signaling pathways. This study, herein, reveals that collagen peptide FNLRMQ could be used as a potential candidate compound for antihypertensive treatment, and could provide scientific evidence for the high-value utilization of marine resources including T. bimaculatus.
Collapse
Affiliation(s)
- Shuilin Cai
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; (S.C.); (Y.S.)
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China; (N.P.); (M.X.); (K.Q.); (B.C.)
| | - Nan Pan
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China; (N.P.); (M.X.); (K.Q.); (B.C.)
| | - Min Xu
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China; (N.P.); (M.X.); (K.Q.); (B.C.)
- College of Food and Biological Engineering, Jimei University, Xiamen 361021, China
| | - Yongchang Su
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; (S.C.); (Y.S.)
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China; (N.P.); (M.X.); (K.Q.); (B.C.)
| | - Kun Qiao
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China; (N.P.); (M.X.); (K.Q.); (B.C.)
| | - Bei Chen
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China; (N.P.); (M.X.); (K.Q.); (B.C.)
| | - Bingde Zheng
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; (S.C.); (Y.S.)
- Correspondence: (B.Z.); (M.X.); (Z.L.)
| | - Meitian Xiao
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China; (S.C.); (Y.S.)
- Correspondence: (B.Z.); (M.X.); (Z.L.)
| | - Zhiyu Liu
- Key Laboratory of Cultivation and High-value Utilization of Marine Organisms in Fujian Province, Fisheries Research Institute of Fujian, Xiamen 361013, China; (N.P.); (M.X.); (K.Q.); (B.C.)
- Correspondence: (B.Z.); (M.X.); (Z.L.)
| |
Collapse
|
31
|
Salvia miltiorrhiza Protects Endothelial Dysfunction against Mitochondrial Oxidative Stress. Life (Basel) 2021; 11:life11111257. [PMID: 34833133 PMCID: PMC8622679 DOI: 10.3390/life11111257] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 11/05/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022] Open
Abstract
Salvia miltiorrhiza (SM) is a common traditional Chinese medicine used in the treatment of cardiovascular and cerebrovascular diseases. Endothelial dysfunction plays an important role in the pathology of cardiovascular diseases. Endothelial dysfunction may induce inflammation and change vascular tone and permeability. The main pathological mechanism of endothelial dysfunction is the formation of reactive oxygen species (ROS). Mitochondria are the main source of energy and can also produce large amounts of ROS. Recent studies have shown that extracts of SM have antioxidative, anti-inflammatory, and antithrombus properties. In this review, we discuss the mechanism of oxidative stress in the mitochondria, endothelial dysfunction, and the role of SM in these oxidative events.
Collapse
|
32
|
Chilosi M, Poletti V, Ravaglia C, Rossi G, Dubini A, Piciucchi S, Pedica F, Bronte V, Pizzolo G, Martignoni G, Doglioni C. The pathogenic role of epithelial and endothelial cells in early-phase COVID-19 pneumonia: victims and partners in crime. Mod Pathol 2021; 34:1444-1455. [PMID: 33883694 PMCID: PMC8058579 DOI: 10.1038/s41379-021-00808-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 02/04/2023]
Abstract
Current understanding of the complex pathogenesis of COVID-19 interstitial pneumonia pathogenesis in the light of biopsies carried out in early/moderate phase and histology data obtained at postmortem analysis is discussed. In autopsies the most observed pattern is diffuse alveolar damage with alveolar-epithelial type-II cell hyperplasia, hyaline membranes, and frequent thromboembolic disease. However, these observations cannot explain some clinical, radiological and physiopathological features observed in SARS-CoV-2 interstitial pneumonia, including the occurrence of vascular enlargement on CT and preserved lung compliance in subjects even presenting with or developing respiratory failure. Histological investigation on early-phase pneumonia on perioperative samples and lung biopsies revealed peculiar morphological and morpho-phenotypical changes including hyper-expression of phosphorylated STAT3 and immune checkpoint molecules (PD-L1 and IDO) in alveolar-epithelial and endothelial cells. These features might explain in part these discrepancies.
Collapse
Affiliation(s)
- Marco Chilosi
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy.
| | - Venerino Poletti
- Department of Respiratory Diseases and Allergy, Aarhus University Hospital, Aarhus, Denmark
- Department of Diseases of the Thorax, G.B. Morgagni Hospital, Forlì, Italy
| | - Claudia Ravaglia
- Department of Diseases of the Thorax, G.B. Morgagni Hospital, Forlì, Italy
| | - Giulio Rossi
- Department of Pathology, Ravenna Hospital, Ravenna, Italy
| | | | - Sara Piciucchi
- Department of Radiology, G.B. Morgagni Hospital, Forlì, Italy
| | - Federica Pedica
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
| | - Vincenzo Bronte
- Department of Medicine, Section of Immunology, University of Verona, Verona, Italy
| | - Giovanni Pizzolo
- Department of Medicine, Section of Hematology, University of Verona, Verona, Italy
| | - Guido Martignoni
- Department of Pathology, Pederzoli Hospital, Peschiera del Garda, Italy
- Department of Pathology and Diagnostics, University of Verona, Verona, Italy
| | - Claudio Doglioni
- Department of Pathology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
33
|
Mallavarpu Ambrose J, Priya Veeraraghavan V, Kullappan M, Chellapandiyan P, Krishna Mohan S, Manivel VA. Comparison of Immunological Profiles of SARS-CoV-2 Variants in the COVID-19 Pandemic Trends: An Immunoinformatics Approach. Antibiotics (Basel) 2021; 10:535. [PMID: 34066389 PMCID: PMC8148159 DOI: 10.3390/antibiotics10050535] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023] Open
Abstract
The current dynamics of the COVID-19 pandemic have become a serious concern with the emergence of a series of mutant variants of the SARS-CoV-2 virus. Unlike the previous strain, it is reported that the descendants are associated with increased risk of transmission yet causing less impact in terms of hospital admission, the severity of illness, or mortality. Moreover, the vaccine efficacy is also not believed to vary among the population depending on the variants of the virus and ethnicity. It has been determined that the mutations recorded in the spike gene and protein of the newly evolved viruses are specificallyresponsible for this transformation in the behavior of the virus and its disease condition. Hence, this study aimed to compare the immunogenic profiles of the spike protein from the latest variants of the SARS-CoV-2 virus concerning the probability of COVID-19 severity. Genome sequences of the latest SARS-CoV-2 variants were obtained from GISAID and NCBI repositories. The translated protein sequences were run against T-cell and B-cell epitope prediction tools. Subsequently, antigenicity, immunogenicity, allergenicity, toxicity, and conservancy of the identified epitopes were ascertained using various prediction servers. Only the non-allergic and non-toxic potential epitopes were matched for population relevance by using the Human Leucocyte Antigen population registry in IEDB. Finally, the selected epitopes were validated by docking and simulation studies. The evaluated immunological parameters would concurrently reveal the severity of COVID-19, determining the infection rate of the pathogen. Our immunoinformatics approach disclosed that spike protein of the five variants was capable of forming potential T and B-cell epitopes with varying immune responses. Although the Wuhan strain showed a high number of epitope/HLA combinations, relatively less antigenicity and higher immunogenicity results in poor neutralizing capacity, which could be associated with increased disease severity. Our data demonstrate that increased viral antigenicity with moderate to high immunogenicity, and several potential epitope/HLA combinations in England strain, the USA, India, and South Africa variants, could possess a high neutralizing ability. Therefore, our findings reinforce that the newly circulating variants of SARS-CoV-2 might be associated with more infectiousness and less severe disease condition despite their greater viremia, as reported in the recent COVID-19 cases, whichconsequently determine their increased epidemiological fitness.
Collapse
Affiliation(s)
- Jenifer Mallavarpu Ambrose
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, Tamil Nadu, India; (J.M.A.); (M.K.)
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University, Velappanchavadi, Chennai 600 077, Tamil Nadu, India;
| | - Malathi Kullappan
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, Tamil Nadu, India; (J.M.A.); (M.K.)
| | - Poongodi Chellapandiyan
- Department of Obstetrics & Gynaecological Nursing, Panimalar College of Nursing, Varadharajapuram, Poonamallee, Chennai 600 123, Tamil Nadu, India;
| | - Surapaneni Krishna Mohan
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, Tamil Nadu, India; (J.M.A.); (M.K.)
- Departments of Biochemistry, Molecular Virology, Clinical Skills and Simulation, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, Tamil Nadu, India
| | - Vivek Anand Manivel
- Department of Research, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600 123, Tamil Nadu, India; (J.M.A.); (M.K.)
- Rudbeck Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, 75185 Uppsala, Sweden
| |
Collapse
|
34
|
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
|
35
|
Secondary Metabolites of Plants as Modulators of Endothelium Functions. Int J Mol Sci 2021; 22:ijms22052533. [PMID: 33802468 PMCID: PMC7959468 DOI: 10.3390/ijms22052533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/20/2021] [Accepted: 02/25/2021] [Indexed: 12/31/2022] Open
Abstract
According to the World Health Organization, cardiovascular diseases are the main cause of death worldwide. They may be caused by various factors or combinations of factors. Frequently, endothelial dysfunction is involved in either development of the disorder or results from it. On the other hand, the endothelium may be disordered for other reasons, e.g., due to infection, such as COVID-19. The understanding of the role and significance of the endothelium in the body has changed significantly over time—from a simple physical barrier to a complex system encompassing local and systemic regulation of numerous processes in the body. Endothelium disorders may arise from impairment of one or more signaling pathways affecting dilator or constrictor activity, including nitric oxide–cyclic guanosine monophosphate activation, prostacyclin–cyclic adenosine monophosphate activation, phosphodiesterase inhibition, and potassium channel activation or intracellular calcium level inhibition. In this review, plants are summarized as sources of biologically active substances affecting the endothelium. This paper compares individual substances and mechanisms that are known to affect the endothelium, and which subsequently may cause the development of cardiovascular disorders.
Collapse
|
36
|
Tran TT, Mai TP, Tran HCB, Le LHG, Vu HA, Tran TK, Hoang SV, Chau HN, Do MD. Association Between AGT M235T and Left Ventricular Mass in Vietnamese Patients Diagnosed With Essential Hypertension. Front Cardiovasc Med 2021; 8:608948. [PMID: 33681303 PMCID: PMC7933009 DOI: 10.3389/fcvm.2021.608948] [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: 09/22/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Background: Increasing left ventricular mass in hypertensive patients is an independent prognostic marker for adverse cardiovascular outcomes. Genetic factors have been shown to critically affect left ventricular mass. AGT M235T is one of the genetic polymorphisms that may influence left ventricular mass due to its pivotal role in the regulation of plasma angiotensinogen level as well as hypertension pathophysiology in Asian populations. Currently, how M235T affects left ventricular mass is not well-described in Vietnamese hypertensive patients. This study aimed to investigate the association between M235T and left ventricular mass in Vietnamese patients diagnosed with essential hypertension. Materials and Methods: AGT M235T genotyping and 2D echocardiography were performed on 187 Vietnamese subjects with essential hypertension. All the ultrasound parameters were obtained to calculate the left ventricular mass index according to the American Society of Echocardiography and the European Association of Cardiovascular Imaging 2015 guidelines. Other clinical characteristics were also recorded, including age, gender, duration of hypertension, hypertensive treatment, lifestyle, renal function, fasting plasma glucose, and lipid profile. Results: MT and TT genotypes were determined in 30 and 157 subjects, respectively. AGT M235T genotype, duration of hypertension, body mass index, and ejection fraction statistically affected the left ventricular mass index, which was significantly greater in TT compared to MT carriers after adjusting for confounding factors. Conclusion: The TT genotype of AGT M23T was associated with greater left ventricular mass in Vietnamese patients diagnosed with essential hypertension.
Collapse
Affiliation(s)
- Tuan Thanh Tran
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Thao Phuong Mai
- Department of Physiology-Pathophysiology-Immunology, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Ha Chau Bich Tran
- Center for Cardiovascular Medicine, University Medical Center, Ho Chi Minh City, Vietnam
| | - Linh Hoang Gia Le
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Hoang Anh Vu
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Trang Kim Tran
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Sy Van Hoang
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Hoa Ngoc Chau
- Department of Internal Medicine, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Minh Duc Do
- Center for Molecular Biomedicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| |
Collapse
|
37
|
Ferrucci R, Dini M, Groppo E, Rosci C, Reitano MR, Bai F, Poletti B, Brugnera A, Silani V, D’Arminio Monforte A, Priori A. Long-Lasting Cognitive Abnormalities after COVID-19. Brain Sci 2021; 11:235. [PMID: 33668456 PMCID: PMC7917789 DOI: 10.3390/brainsci11020235] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/03/2021] [Accepted: 02/09/2021] [Indexed: 02/07/2023] Open
Abstract
Considering the mechanisms capable of causing brain alterations in COVID-19, we aimed to study the occurrence of cognitive abnormalities in the months following hospital discharge. We recruited 38 (aged 22-74 years; 27 males) patients hospitalized for complications of SARS-CoV-2 infection in nonintensive COVID units. Participants underwent neuropsychological testing about 5 months after hospital discharge. Of all patients, 42.1% had processing speed deficits, while 26.3% showed delayed verbal recall deficits. Twenty-one percent presented with deficits in both processing speed and verbal memory. Bivariate analysis revealed a positive correlation between the lowest arterial oxygen partial pressure (PaO2) to fractional inspired oxygen (FiO2) (P/F) ratio during hospitalization and verbal memory consolidation performance (SRT-LTS score, r = 0.404, p = 0.027), as well as a positive correlation between SpO2 levels upon hospital arrival and delayed verbal recall performance (SRT-D score, rs = 0.373, p = 0.042). Acute respiratory distress syndrome (ARDS) during hospitalization was associated with worse verbal memory performance (ARDS vs. no ARDS: SRT-LTS mean score = 30.63 ± 13.33 vs. 44.50 ± 13.16, p = 0.007; SRT-D mean score = 5.95 ± 2.56 vs. 8.10 ± 2.62, p = 0.029). Cognitive abnormalities can frequently be found in COVID-19 patients 5 months after hospital discharge. Increased fatigability, deficits of concentration and memory, and overall decreased cognitive speed months after hospital discharge can interfere with work and daily activities.
Collapse
Affiliation(s)
- Roberta Ferrucci
- Aldo Ravelli Research Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, 20142 Milan, Italy; (R.F.); (M.D.); (V.S.)
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
- Department of Health Science (DISS), University of Milan, 20142 Milan, Italy
| | - Michelangelo Dini
- Aldo Ravelli Research Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, 20142 Milan, Italy; (R.F.); (M.D.); (V.S.)
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
- Department of Health Science (DISS), University of Milan, 20142 Milan, Italy
| | - Elisabetta Groppo
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
| | - Chiara Rosci
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
| | - Maria Rita Reitano
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
| | - Francesca Bai
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
- Department of Health Science (DISS), University of Milan, 20142 Milan, Italy
| | - Barbara Poletti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italian, 20149 Milan, Italy;
| | - Agostino Brugnera
- Department of Human and Social sciences, University of Bergamo, 24129 Bergamo, Italy;
| | - Vincenzo Silani
- Aldo Ravelli Research Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, 20142 Milan, Italy; (R.F.); (M.D.); (V.S.)
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italian, 20149 Milan, Italy;
- Department of Pathophysiology and Transplantation, “Dino Ferrari” Center, University of Milano, 20122 Milan, Italy
| | - Antonella D’Arminio Monforte
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
- Department of Health Science (DISS), University of Milan, 20142 Milan, Italy
| | - Alberto Priori
- Aldo Ravelli Research Center for Neurotechnology and Experimental Brain Therapeutics, University of Milan, 20142 Milan, Italy; (R.F.); (M.D.); (V.S.)
- ASST-Santi Paolo e Carlo University Hospital, 20142 Milan, Italy; (E.G.); (C.R.); (M.R.R.); (F.B.); (A.D.M.)
- Department of Health Science (DISS), University of Milan, 20142 Milan, Italy
| |
Collapse
|
38
|
Forsyth KS, Anguera MC. Time to get ill: the intersection of viral infections, sex, and the X chromosome. CURRENT OPINION IN PHYSIOLOGY 2021; 19:62-72. [PMID: 33073073 PMCID: PMC7553007 DOI: 10.1016/j.cophys.2020.09.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Females have more robust immune responses than males, and viral infections are more severe for males. Hormones and genetic sex, namely the X chromosome, influence sex differences with immune responses. Here, we review recent findings underlying sexual dimorphism of disease susceptibility for two prevalent viral infections, influenza and SARS-CoV-2, which exhibit male-biased disease severity. Viral infections are proposed to be an initiating event for autoimmunity, which exhibits a female bias. We also review recent work elucidating the epigenetic and genetic contribution of X-Chromosome Inactivation maintenance, and X-linked gene expression, for the autoimmune disorder Systemic Lupus Erythematosus, and highlight the complex considerations required for identifying underlying hormonal and genetic contributions responsible for sex differences in immune responses.
Collapse
Affiliation(s)
- Katherine S Forsyth
- Dept. of Biomedical Sciences, University of Pennsylvania, Philadelphia PA 19104, United States
| | - Montserrat C Anguera
- Dept. of Biomedical Sciences, University of Pennsylvania, Philadelphia PA 19104, United States
| |
Collapse
|
39
|
Hadzhibozheva P, Tolekova A, Kalfin R, Georgiev T. Analysis of angiotensin II-Induced rat urinary bladder contractions in the presence of angiotensin II receptors blockers. Arch Physiol Biochem 2021; 127:1-5. [PMID: 30739509 DOI: 10.1080/13813455.2018.1555669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
OBJECTIVE An application of a specific analysis on recordings obtained from urinary bladder (UB) preparations influenced with Angiotensin II (AngII) and AngII receptor (ATR) blockers was performed. METHODS UB preparations were divided as follows: group 1 stimulated with AngII only; group 2:PD123319 (ATR type-2 blocker)+AngII; group 3:Losartan (ATR type-1 blocker)+AngII. The averaged time and force parameters of the contractions were processed by a spline interpolation and graphic images of the different patterns of the contractile activity were obtained. RESULTS The speed of AngII-induced UB contraction, when PD123319 was administered, was significantly higher than those, registered by the application of AngII alone and Losartan + AngII. The presence of Losartan markedly delayed the speed of the overall AngII-induced contraction. CONCLUSION The study indicates the contribution of both ATR subtypes for the development of AngII-induced UB contraction. Our results showed that probably ATR mediate a reciprocal dynamic response to AngII in the bladder.
Collapse
Affiliation(s)
- Petya Hadzhibozheva
- Faculty of Medicine, Department of Physiology, Pathophysiology and Pharmacology, Trakia University, Stara Zagora, Bulgaria
| | - Anna Tolekova
- Faculty of Medicine, Department of Physiology, Pathophysiology and Pharmacology, Trakia University, Stara Zagora, Bulgaria
| | - Reni Kalfin
- Institute of Neurobiology, Bulgarian Academy of Scienses, Sofia, Bulgaria
| | - Tsvetelin Georgiev
- Faculty of Medicine, Department of Physiology, Pathophysiology and Pharmacology, Trakia University, Stara Zagora, Bulgaria
| |
Collapse
|
40
|
Cheruiyot I, Kipkorir V, Ngure B, Misiani M, Munguti J, Ogeng'o J. Arterial Thrombosis in Coronavirus Disease 2019 Patients: A Rapid Systematic Review. Ann Vasc Surg 2021; 70:273-281. [PMID: 32866574 PMCID: PMC7453204 DOI: 10.1016/j.avsg.2020.08.087] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 08/16/2020] [Indexed: 12/23/2022]
Abstract
BACKGROUND Emerging evidence suggests that severe form of coronavirus disease 2019 (COVID-19) is mediated, in part, by a hypercoagulable state characterized by micro- and macro-vascular thrombotic angiopathy. Although venous thrombotic events in COVID-19 patients have been well described, data on arterial thrombosis (AT) in these patients is still limited. We, therefore, conducted a rapid systematic review of current scientific literature to identify and consolidate evidence of AT in COVID-19 patients. METHODS A systematic search of literature was conducted between November 1, 2019, and June 9, 2020, on PubMed and China National Knowledge Infrastructure to identify potentially eligible studies. RESULTS A total of 27 studies (5 cohort, 5 case series, and 17 case reports) describing arterial thrombotic events in 90 COVID-19 patients were included. The pooled incidence of AT in severe/critically ill intensive care unit-admitted COVID-19 patients across the 5 cohort studies was 4.4% (95% confidence interval 2.8-6.4). Most of the patients were male, elderly, and had comorbidities. AT was symptomatic in >95% of these patients and involved multiple arteries in approximately 18% of patients. The anatomical distribution of arterial thrombotic events was wide, occurring in limb arteries (39%), cerebral arteries (24%), great vessels (aorta, common iliac, common carotid, and brachiocephalic trunk; 19%), coronary arteries (9%), and superior mesenteric artery (8%). The mortality rate in these patients is approximately 20%. CONCLUSIONS AT occurs in approximately 4% of critically ill COVID-19 patients. It often presents symptomatically and can affect multiple arteries. Further investigation of the underlying mechanism of AT in COVID-19 would be needed to clarify possible therapeutic targets.
Collapse
Affiliation(s)
- Isaac Cheruiyot
- Department of Human Anatomy, School of Medicine, University of Nairobi, Nairobi, Kenya.
| | - Vincent Kipkorir
- Department of Human Anatomy, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Brian Ngure
- Department of Human Anatomy, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Musa Misiani
- Department of Human Anatomy, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Jeremiah Munguti
- Department of Human Anatomy, School of Medicine, University of Nairobi, Nairobi, Kenya
| | - Julius Ogeng'o
- Department of Human Anatomy, School of Medicine, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
41
|
COVID-19 and thrombosis: From bench to bedside. Trends Cardiovasc Med 2020; 31:143-160. [PMID: 33338635 PMCID: PMC7836332 DOI: 10.1016/j.tcm.2020.12.004] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 12/12/2020] [Accepted: 12/12/2020] [Indexed: 12/13/2022]
Abstract
Coronavirus disease of 2019 (COVID-19) is the respiratory viral infection caused by the coronavirus SARS-CoV2 (Severe Acute Respiratory Syndrome Coronavirus 2). Despite being a respiratory illness, COVID-19 is found to increase the risk of venous and arterial thromboembolic events. Indeed, the link between COVID-19 and thrombosis is attracting attention from the broad scientific community. In this review we will analyze the current available knowledge of the association between COVID-19 and thrombosis. We will highlight mechanisms at both molecular and cellular levels that may explain this association. In addition, the article will review the antithrombotic properties of agents currently utilized or being studied in COVID-19 management. Finally, we will discuss current professional association guidance on prevention and treatment of thromboembolism associated with COVID-19.
Collapse
|
42
|
ACE2, angiotensin 1-7 and skeletal muscle: review in the era of COVID-19. Clin Sci (Lond) 2020; 134:3047-3062. [PMID: 33231620 PMCID: PMC7687025 DOI: 10.1042/cs20200486] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiotensin converting enzyme-2 (ACE2) is a multifunctional transmembrane protein recently recognised as the entry receptor of the virus causing COVID-19. In the renin–angiotensin system (RAS), ACE2 cleaves angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7), which is considered to exert cellular responses to counteract the activation of the RAS primarily through a receptor, Mas, in multiple organs including skeletal muscle. Previous studies have provided abundant evidence suggesting that Ang 1-7 modulates multiple signalling pathways leading to protection from pathological muscle remodelling and muscle insulin resistance. In contrast, there is relatively little evidence to support the protective role of ACE2 in skeletal muscle. The potential contribution of endogenous ACE2 to the regulation of Ang 1-7-mediated protection of these muscle pathologies is discussed in this review. Recent studies have suggested that ACE2 protects against ageing-associated muscle wasting (sarcopenia) through its function to modulate molecules outside of the RAS. Thus, the potential association of sarcopenia with ACE2 and the associated molecules outside of RAS is also presented herein. Further, we introduce the transcriptional regulation of muscle ACE2 by drugs or exercise, and briefly discuss the potential role of ACE2 in the development of COVID-19.
Collapse
|
43
|
Nejat R, Sadr AS. Are losartan and imatinib effective against SARS-CoV2 pathogenesis? A pathophysiologic-based in silico study. In Silico Pharmacol 2020; 9:1. [PMID: 33294307 PMCID: PMC7716628 DOI: 10.1007/s40203-020-00058-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022] Open
Abstract
Proposing a theory about the pathophysiology of cytokine storm in COVID19, we were to find the potential drugs to treat this disease and to find any effect of these drugs on the virus infectivity through an in silico study. COVID-19-induced ARDS is linked to a cytokine storm phenomenon not explainable solely by the virus infectivity. Knowing that ACE2, the hydrolyzing enzyme of AngII and SARS-CoV2 receptor, downregulates when the virus enters the host cells, we hypothesize that hyperacute AngII upregulation is the eliciting factor of this ARDS. We were to validate this theory through reviewing previous studies to figure out the role of overzealous activation of AT1R in ARDS. According to this theory losartan may attenuate ARDS in this disease. Imatinib, has previously been elucidated to be promising in modulating lung inflammatory reactions and virus infectivity in SARS and MERS. We did an in silico study to uncover any probable other unconsidered inhibitory effects of losartan and imatinib against SARS-CoV2 pathogenesis. Reviewing the literature, we could find that over-activation of AT1R could explain precisely the mechanism of cytokine storm in COVID19. Our in silico study revealed that losartan and imatinib could probably: (1) decline SARS-CoV2 affinity to ACE2. (2) inhibit the main protease and furin, (3) disturb papain-like protease and p38MAPK functions. Our reviewing on renin-angiotensin system showed that overzealous activation of AT1R by hyper-acute excess of AngII due to acute downregulation of ACE2 by SARS-CoV2 explains precisely the mechanism of cytokine storm in COVID-19. Besides, based on our in silico study we concluded that losartan and imatinib are promising in COVID19.
Collapse
Affiliation(s)
- Reza Nejat
- Department of Anesthesiology and Critical Care Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ahmad Shahir Sadr
- Bioinformatics Research Center, Cheragh Medical Institute and Hospital, Kabul, Afghanistan
- Department of Computer Science, Faculty of Mathematical Sciences, Shahid Beheshti University, Tehran, Iran
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| |
Collapse
|
44
|
Del Turco S, Vianello A, Ragusa R, Caselli C, Basta G. COVID-19 and cardiovascular consequences: Is the endothelial dysfunction the hardest challenge? Thromb Res 2020; 196:143-151. [PMID: 32871306 PMCID: PMC7451195 DOI: 10.1016/j.thromres.2020.08.039] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 08/17/2020] [Accepted: 08/26/2020] [Indexed: 12/22/2022]
Abstract
A Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2) has become a pandemic disease named Coronavirus Disease-19 (COVID-19) of epochal dimension. The clinical spectrum of COVID-19 is wide, ranging from asymptomatic forms to severe pneumonia, sepsis and multiple organ dysfunction syndromes resulting in poor outcomes. Among the various consequences of severe COVID-19, cardiovascular (CV) collapse appears the most serious and potentially lethal. On the other hand, pre-existent CV comorbidities are also associated with higher mortality. The most reliable hypothetical pathogenetic mechanism for CV complications and cardiac injury in severe COVID-19 patients appears to be a sustained endothelial dysfunction, caused by the interplay of inflammation and coagulation. In this review, we survey papers addressing issues related to severe COVID-19, characterized by enhanced lung microvascular loss, hypercytokinemia, hypoxemia and thrombosis. We discuss about how the virus-induced downregulation of the angiotensin converting enzyme-2 (ACE2) receptor, used to enter the host cell, could affect the renin-angiotensin system, attempting to clarify the doubts about the use of ACE inhibitors and Angiotensin-II receptor blockers in COVID-19 patients. Finally, we point out how the delicate and physiological homeostatic function of the endothelium, which turns into a disastrous battlefield of the complex interaction between "cytokine and coagulative storms", can be irreparably compromised and result in systemic inflammatory complications.
Collapse
Affiliation(s)
- Serena Del Turco
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy.
| | - Annamaria Vianello
- Department of Information Engineering, Telemedicine Section, University of Pisa, Italy
| | - Rosetta Ragusa
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy
| | - Chiara Caselli
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy
| | - Giuseppina Basta
- Institute of Clinical Physiology, CNR, San Cataldo Research Area, Via Moruzzi, 1, 56124 Pisa, Italy.
| |
Collapse
|
45
|
Song L, Chen X, Swanson TA, LaViolette B, Pang J, Cunio T, Nagle MW, Asano S, Hales K, Shipstone A, Sobon H, Al-Harthy SD, Ahn Y, Kreuser S, Robertson A, Ritenour C, Voigt F, Boucher M, Sun F, Sessa WC, Roth Flach RJ. Lymphangiogenic therapy prevents cardiac dysfunction by ameliorating inflammation and hypertension. eLife 2020; 9:e58376. [PMID: 33200983 PMCID: PMC7695461 DOI: 10.7554/elife.58376] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 11/16/2020] [Indexed: 12/11/2022] Open
Abstract
The lymphatic vasculature is involved in the pathogenesis of acute cardiac injuries, but little is known about its role in chronic cardiac dysfunction. Here, we demonstrate that angiotensin II infusion induced cardiac inflammation and fibrosis at 1 week and caused cardiac dysfunction and impaired lymphatic transport at 6 weeks in mice, while co-administration of VEGFCc156s improved these parameters. To identify novel mechanisms underlying this protection, RNA sequencing analysis in distinct cell populations revealed that VEGFCc156s specifically modulated angiotensin II-induced inflammatory responses in cardiac and peripheral lymphatic endothelial cells. Furthermore, telemetry studies showed that while angiotensin II increased blood pressure acutely in all animals, VEGFCc156s-treated animals displayed a delayed systemic reduction in blood pressure independent of alterations in angiotensin II-mediated aortic stiffness. Overall, these results demonstrate that VEGFCc156s had a multifaceted therapeutic effect to prevent angiotensin II-induced cardiac dysfunction by improving cardiac lymphatic function, alleviating fibrosis and inflammation, and ameliorating hypertension.
Collapse
Affiliation(s)
- LouJin Song
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Xian Chen
- Comparative Medicine, Pfizer IncCambridgeUnited States
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Terri A Swanson
- Early Clinical Development, Pfizer IncCambridgeUnited States
| | | | - Jincheng Pang
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Teresa Cunio
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
- Acceleron PharmaCambridgeUnited States
| | - Michael W Nagle
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
- Eisai IncCambridgeUnited States
| | - Shoh Asano
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Katherine Hales
- Internal Medicine Research Unit, Pfizer IncCambridgeUnited States
| | - Arun Shipstone
- Inflammation and Immunology Research Unit, Pfizer IncCambridgeUnited States
| | - Hanna Sobon
- Inflammation and Immunology Research Unit, Pfizer IncCambridgeUnited States
| | - Sabra D Al-Harthy
- Comparative Medicine, Pfizer IncCambridgeUnited States
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Youngwook Ahn
- Target Sciences, Emerging Science and Innovation, Pfizer IncCambridgeUnited States
| | | | - Andrew Robertson
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Casey Ritenour
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Frank Voigt
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Magalie Boucher
- Drug Safety Research & Development, Pfizer IncGrotonUnited States
| | - Furong Sun
- Early Clinical Development, Pfizer IncCambridgeUnited States
| | - William C Sessa
- Department of Pharmacology, Vascular Biology and Therapeutics Program, Yale University School of MedicineNew HavenUnited States
| | | |
Collapse
|
46
|
Sriram K, Insel PA. A hypothesis for pathobiology and treatment of COVID-19: The centrality of ACE1/ACE2 imbalance. Br J Pharmacol 2020; 177:4825-4844. [PMID: 32333398 PMCID: PMC7572451 DOI: 10.1111/bph.15082] [Citation(s) in RCA: 120] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 11/29/2022] Open
Abstract
Angiotensin Converting Enzyme2 is the cell surface binding site for the coronavirus SARS-CoV-2, which causes COVID-19. We propose that an imbalance in the action of ACE1- and ACE2-derived peptides, thereby enhancing angiotensin II (Ang II) signalling is primary driver of COVID-19 pathobiology. ACE1/ACE2 imbalance occurs due to the binding of SARS-CoV-2 to ACE2, reducing ACE2-mediated conversion of Ang II to Ang peptides that counteract pathophysiological effects of ACE1-generated ANG II. This hypothesis suggests several approaches to treat COVID-19 by restoring ACE1/ACE2 balance: (a) AT receptor antagonists; (b) ACE1 inhibitors (ACEIs); (iii) agonists of receptors activated by ACE2-derived peptides (e.g. Ang (1-7), which activates MAS1); (d) recombinant human ACE2 or ACE2 peptides as decoys for the virus. Reducing ACE1/ACE2 imbalance is predicted to blunt COVID-19-associated morbidity and mortality, especially in vulnerable patients. Importantly, approved AT antagonists and ACEIs can be rapidly repurposed to test their efficacy in treating COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of PharmacologyUniversity of California San DiegoLa JollaCAUSA
| | - Paul A. Insel
- Department of PharmacologyUniversity of California San DiegoLa JollaCAUSA
- Department of MedicineUniversity of California San DiegoLa JollaCAUSA
| |
Collapse
|
47
|
Sriram K, Insel PA. A hypothesis for pathobiology and treatment of COVID-19: The centrality of ACE1/ACE2 imbalance. Br J Pharmacol 2020. [PMID: 32333398 DOI: 10.1111/bph.15082.10.1111/bph.15082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/21/2023] Open
Abstract
Angiotensin Converting Enzyme2 is the cell surface binding site for the coronavirus SARS-CoV-2, which causes COVID-19. We propose that an imbalance in the action of ACE1- and ACE2-derived peptides, thereby enhancing angiotensin II (Ang II) signalling is primary driver of COVID-19 pathobiology. ACE1/ACE2 imbalance occurs due to the binding of SARS-CoV-2 to ACE2, reducing ACE2-mediated conversion of Ang II to Ang peptides that counteract pathophysiological effects of ACE1-generated ANG II. This hypothesis suggests several approaches to treat COVID-19 by restoring ACE1/ACE2 balance: (a) AT receptor antagonists; (b) ACE1 inhibitors (ACEIs); (iii) agonists of receptors activated by ACE2-derived peptides (e.g. Ang (1-7), which activates MAS1); (d) recombinant human ACE2 or ACE2 peptides as decoys for the virus. Reducing ACE1/ACE2 imbalance is predicted to blunt COVID-19-associated morbidity and mortality, especially in vulnerable patients. Importantly, approved AT antagonists and ACEIs can be rapidly repurposed to test their efficacy in treating COVID-19. LINKED ARTICLES: This article is part of a themed issue on The Pharmacology of COVID-19. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.21/issuetoc.
Collapse
Affiliation(s)
- Krishna Sriram
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
| | - Paul A Insel
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
48
|
Sparks MA, South AM, Badley AD, Baker-Smith CM, Batlle D, Bozkurt B, Cattaneo R, Crowley SD, Dell’Italia LJ, Ford AL, Griendling K, Gurley SB, Kasner SE, Murray JA, Nath KA, Pfeffer MA, Rangaswami J, Taylor WR, Garovic VD. Severe Acute Respiratory Syndrome Coronavirus 2, COVID-19, and the Renin-Angiotensin System: Pressing Needs and Best Research Practices. Hypertension 2020; 76:1350-1367. [PMID: 32981369 PMCID: PMC7685174 DOI: 10.1161/hypertensionaha.120.15948] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is associated with significant morbidity and mortality throughout the world, predominantly due to lung and cardiovascular injury. The virus responsible for COVID-19-severe acute respiratory syndrome coronavirus 2-gains entry into host cells via ACE2 (angiotensin-converting enzyme 2). ACE2 is a primary enzyme within the key counter-regulatory pathway of the renin-angiotensin system (RAS), which acts to oppose the actions of Ang (angiotensin) II by generating Ang-(1-7) to reduce inflammation and fibrosis and mitigate end organ damage. As COVID-19 spans multiple organ systems linked to the cardiovascular system, it is imperative to understand clearly how severe acute respiratory syndrome coronavirus 2 may affect the multifaceted RAS. In addition, recognition of the role of ACE2 and the RAS in COVID-19 has renewed interest in its role in the pathophysiology of cardiovascular disease in general. We provide researchers with a framework of best practices in basic and clinical research to interrogate the RAS using appropriate methodology, especially those who are relatively new to the field. This is crucial, as there are many limitations inherent in investigating the RAS in experimental models and in humans. We discuss sound methodological approaches to quantifying enzyme content and activity (ACE, ACE2), peptides (Ang II, Ang-[1-7]), and receptors (types 1 and 2 Ang II receptors, Mas receptor). Our goal is to ensure appropriate research methodology for investigations of the RAS in patients with severe acute respiratory syndrome coronavirus 2 and COVID-19 to ensure optimal rigor and reproducibility and appropriate interpretation of results from these investigations.
Collapse
Affiliation(s)
- Matthew A. Sparks
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC
- Renal Section, Durham VA Health Care System, Durham, NC
- American Heart Association, Council on Kidney in Cardiovascular Disease
| | - Andrew M. South
- American Heart Association, Council on Kidney in Cardiovascular Disease
- American Heart Association, Council on Hypertension
- Section of Nephrology, Department of Pediatrics, Brenner Children’s Hospital, Wake Forest School of Medicine, Winston Salem, NC
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston Salem, NC
- Department of Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine, Winston Salem, NC
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston Salem, NC
| | - Andrew D. Badley
- Division of Infectious Diseases, Mayo Clinic College of Medicine, Rochester, MN
| | - Carissa M. Baker-Smith
- Director of Preventive Cardiology, Division of Pediatric Cardiology, Department of Pediatrics, Nemours/Alfred I. duPont Hospital for Children, Wilmington, DE
- American Heart Association, Council on Lifelong Congenital Heart Disease and Heart Health in the Young
| | - Daniel Batlle
- Division of Nephrology and Hypertension, Northwestern University Feinberg Medical School, Chicago, IL
- American Heart Association, Council on Hypertension
| | - Biykem Bozkurt
- Section of Cardiology, Department of Internal Medicine, Baylor College of Medicine, Houston, TX
- Michael E. DeBakey VA Medical Center, Houston, TX
- American Heart Association, Council on Clinical Cardiology
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic College of Medicine, Rochester, MN
| | - Steven D. Crowley
- Division of Nephrology, Department of Medicine, Duke University School of Medicine, Durham, NC
- Renal Section, Durham VA Health Care System, Durham, NC
- American Heart Association, Council on Kidney in Cardiovascular Disease
| | - Louis J. Dell’Italia
- Department of Medicine, Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, AL
- Department of Veterans Affairs Medical Center, Birmingham, AL
- American Heart Association, Council on Basic Cardiovascular Sciences
| | - Andria L. Ford
- Department of Neurology, Washington University in St. Louis School of Medicine, St. Louis, MO
- American Heart Association, Stroke Council
| | - Kathy Griendling
- American Heart Association, Council on Basic Cardiovascular Sciences
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA
| | - Susan B. Gurley
- American Heart Association, Council on Kidney in Cardiovascular Disease
- Department of Medicine, Division of Nephrology and Hypertension, Oregon Health & Science University, Portland, OR
| | - Scott E. Kasner
- Department of Neurology, University of Pennsylvania, Philadelphia, Pennsylvania
- American Heart Association, Stroke Council
| | - Joseph A. Murray
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, MN
| | - Karl A. Nath
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, MN
| | - Marc A. Pfeffer
- American Heart Association, Council on Clinical Cardiology
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Janani Rangaswami
- American Heart Association, Council on Kidney in Cardiovascular Disease
- Department of Medicine, Einstein Medical Center Philadelphia, Philadelphia, PA
- Sidney Kimmel College of Thomas Jefferson University, Philadelphia, PA
| | - W. Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, GA
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA
- Division of Cardiology, Atlanta VA Medical Center, Decatur, GA
- American Heart Association, Council on Arteriosclerosis, Thrombosis and Vascular Biology
| | - Vesna D. Garovic
- American Heart Association, Council on Hypertension
- Division of Nephrology and Hypertension, Mayo Clinic College of Medicine, Rochester, MN
- Department of Obstetrics and Gynecology, Mayo Clinic College of Medicine, Rochester, MN
| |
Collapse
|
49
|
Czick M, Shapter C, Shapter R. COVID's Razor: RAS Imbalance, the Common Denominator Across Disparate, Unexpected Aspects of COVID-19. Diabetes Metab Syndr Obes 2020; 13:3169-3192. [PMID: 32982349 PMCID: PMC7495349 DOI: 10.2147/dmso.s265518] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/04/2020] [Indexed: 12/25/2022] Open
Abstract
A modern iteration of Occam's Razor posits that "the simplest explanation is usually correct." Coronavirus Disease 2019 involves widespread organ damage and uneven mortality demographics, deemed unexpected from what was originally thought to be "a straightforward respiratory virus." The simplest explanation is that both the expected and unexpected aspects of COVID-19 share a common mechanism. Silent hypoxia, atypical acute respiratory distress syndrome (ARDS), stroke, olfactory loss, myocarditis, and increased mortality rates in the elderly, in men, in African-Americans, and in patients with obesity, diabetes, and cancer-all bear the fingerprints of the renin-angiotensin system (RAS) imbalance, suggesting that RAS is the common culprit. This article examines what RAS is and how it works, then from that baseline, the article presents the evidence suggesting RAS involvement in the disparate manifestations of COVID-19. Understanding the deeper workings of RAS helps one make sense of severe COVID-19. In addition, recognizing the role of RAS imbalance suggests potential routes to mitigate COVID-19 severity.
Collapse
Affiliation(s)
- Maureen Czick
- University of Connecticut, Department of Anesthesia, Farmington, CT, USA
| | | | - Robert Shapter
- Independent Consultant ( Medical Research, Medical Communications, and Medical Education), Hartford, CT, USA
| |
Collapse
|
50
|
Abstract
Abstract
At the end of 2019, a new coronavirus infection occurred in the People’s Republic of China with an epicentre in the city of Wuhan. On February 11th, 2020, the World Health Organization assigned the official name of the infection caused by the new coronavirus – COVID-19. COVID-19 has affected people from all over the world given that the infection was noted in 200 countries resulting in annunciation of the pandemic situation. Human corona viruses cause mild to moderate respiratory infections. At the end of 2002, a new coronavirus appeared (SARS-CoV), the causal agent of atypical pneumonia, which caused acute respiratory distress syndrome (ARDS). The initial stage of COVID-19 infection is the penetration of SARS-CoV-2 into target cells that have angiotensin converting enzyme type II receptors. The virus enters the body through the respiratory tract and interacts primarily with toll-like receptors (TLRs). The events in SARS-Cov-2 induced infection follow the next scenario: epithelial cells via TLRs recognize and identify SARS-Cov-2, and after that the information is transmitted to the transcriptional NF-κB, which causes expression of the corresponding genes. Activated in this way, the epithelial cells begin to synthesize various biologically active molecules. The results obtained on preclinical material indicate that ROS generation increases and the antioxidant protection decreases, which plays a major role in the pathogenesis of SARS-CoV, as well as in the progression and severity of this respiratory disease.
Collapse
|