1
|
Tang MY, Xie H, Tao JT, Zhang C, Luo YH, Zhang C, Peng SQ, Xie LX, Lv WB, Zhang C, Huang L. Pathophysiological relevance and therapeutic outlook of GPR43 in atherosclerosis. Biochem Cell Biol 2024; 102:418-429. [PMID: 39013204 DOI: 10.1139/bcb-2024-0053] [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] [Indexed: 07/18/2024] Open
Abstract
Atherosclerosis (AS) is an inflammatory arterial disorder that occurs due to the deposition of the excessive lipoprotein under the artery intima, mainly including low-density lipoprotein and other apolipoprotein B-containing lipoproteins. G protein-coupled receptors (GPCRs) play a crucial role in transmitting signals in physiological and pathophysiological conditions. GPCRs recognize inflammatory mediators, thereby serving as important players during chronic inflammatory processes. It has been demonstrated that free fatty acids can function as ligands for various GPCRs, such as free fatty acid receptor (FFAR)1/GPR40, FFAR2/GPR43, FFAR3/GPR41, FFAR4/GPR120, and the lipid metabolite binding glucose-dependent insulinotropic receptor (GPR119). This review discusses GPR43 and its ligands in the pathogenesis of AS, especially focusing on its distinct role in regulating chronic vascular inflammation, inhibiting oxidative stress, ameliorating endothelial dysfunction and improving dyslipidemia. It is hoped that this review may provide guidance for further studies aimed at GPR43 as a promising target for drug development in the prevention and therapy of AS.
Collapse
Affiliation(s)
- Mu-Yao Tang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Hao Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Jin-Tao Tao
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chun Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Yao-Hua Luo
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Cong Zhang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Si-Qin Peng
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Lin-Xi Xie
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Wen-Bo Lv
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
- Departments of Clinical Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Chi Zhang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| | - Liang Huang
- Research Laboratory of Translational Medicine, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, People's Republic of China
| |
Collapse
|
2
|
Ghasempour Dabaghi G, Rabiee Rad M, Amani-Beni R, Darouei B. The role of p130Cas/BCAR1 adaptor protein in the pathogenesis of cardiovascular diseases: A literature review. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 44:100416. [PMID: 39036012 PMCID: PMC11259988 DOI: 10.1016/j.ahjo.2024.100416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 05/22/2024] [Accepted: 06/23/2024] [Indexed: 07/23/2024]
Abstract
Breast cancer anti-estrogen resistance-1 (p130Cas/BCAR1) is an adaptor protein of the cas(Cas) family. This protein regulates multiple complex pathways in different organs including bones, pancreas, and immune and cardiovascular systems. Although previous research well demonstrated the role of p130Cas/BCAR1 in different diseases especially cancers, a precise review study on the various effects of p130Cas/BCAR1 on cardiovascular diseases is missing. In this study, we reviewed mechanisms of action for p130Cas/BCAR1 impact, on cardiac embryonic development defects, hypertrophy and remodeling, pulmonary artery hypertension (PAH), and atherosclerosis. Also, we suggest feature direction for research and potential therapeutic implications. This study showed that p130Cas/BCAR1 can affect cardiovascular diseases in various mechanisms including actin stress fiber formation, attachment to focal adhesion kinase (FAK) and angiotensin II (Ang II), generation of reactive oxygen species (ROS), and growth factor signaling through amplifying receptor tyrosine kinase (RTKs).
Collapse
Affiliation(s)
- Ghazal Ghasempour Dabaghi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehrdad Rabiee Rad
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Reza Amani-Beni
- School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Bahar Darouei
- School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| |
Collapse
|
3
|
Jin S, Kang PM. A Systematic Review on Advances in Management of Oxidative Stress-Associated Cardiovascular Diseases. Antioxidants (Basel) 2024; 13:923. [PMID: 39199169 PMCID: PMC11351257 DOI: 10.3390/antiox13080923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/10/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
Oxidative stress plays a significant role in the pathogenesis of cardiovascular diseases, such as myocardial ischemia/reperfusion injury, atherosclerosis, heart failure, and hypertension. This systematic review aims to integrate most relevant studies on oxidative stress management in cardiovascular diseases. We searched relevant literatures in the PubMed database using specific keywords. We put emphasis on those manuscripts that were published more recently and in higher impact journals. We reviewed a total of 200 articles. We examined current oxidative stress managements in cardiovascular diseases, including supplements like resveratrol, vitamins C and E, omega-3 fatty acids, flavonoids, and coenzyme-10, which have shown antioxidative properties and potential cardiovascular benefits. In addition, we reviewed the pharmacological treatments including newly discovered antioxidants and nanoparticles that show potential effects in targeting the specific oxidative stress pathways. Lastly, we examined biomarkers, such as soluble transferrin receptor, transthyretin, and cystatin C in evaluating antioxidant status and identifying cardiovascular risk. By addressing oxidative stress management and mechanisms, this paper emphasizes the importance of maintaining the balance between oxidants and antioxidants in the progression of cardiovascular diseases. This review paper is registered with the International Platform of Registered Systematic Review and Meta-analysis Protocols (INPLASY), registration # INPLASY202470064.
Collapse
Affiliation(s)
- Soyeon Jin
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS 910, Boston, MA 02215, USA
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| | - Peter M. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle, CLS 910, Boston, MA 02215, USA
| |
Collapse
|
4
|
Moore GJ, Ridway H, Gadanec LK, Apostolopoulos V, Zulli A, Swiderski J, Kelaidonis K, Vidali VP, Matsoukas MT, Chasapis CT, Matsoukas JM. Structural Features Influencing the Bioactive Conformation of Angiotensin II and Angiotensin A: Relationship between Receptor Desensitization, Addiction, and the Blood-Brain Barrier. Int J Mol Sci 2024; 25:5779. [PMID: 38891966 PMCID: PMC11171751 DOI: 10.3390/ijms25115779] [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: 03/27/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 06/21/2024] Open
Abstract
The N-terminal portion of the octapeptide angiotensin II (DRVYIHPF; AngII), a vasopressor peptide that favorably binds to, and activates, AngII type 1 receptor (AT1R), has an important role in maintaining bioactive conformation. It involves all three charged groups, namely (i) the N-terminal amino group cation, (ii) the Asp sidechain anion and (iii) the Arg guanidino cation. Neutralization of any one of these three charged groups results in a substantial reduction (<5%) in bioactivity, implicating a specialized function for this cluster. In contrast, angiotensin A (ARVYIHPF; AngA) has reduced bioactivity at AT1R; however, replacement of Asp in AngII with sarcosine (N-methyl-glycine) not only restores bioactivity but increases the activity of agonist, antagonist, and inverse agonist analogues. A bend produced at the N-terminus by the introduction of the secondary amino acid sarcosine is thought to realign the functional groups that chaperone the C-terminal portion of AngII, allowing transfer of the negative charge originating at the C-terminus to be transferred to the Tyr hydroxyl-forming tyrosinate anion, which is required to activate the receptor and desensitizes the receptor (tachyphylaxis). Peptide (sarilesin) and nonpeptide (sartans) moieties, which are long-acting inverse agonists, appear to desensitize the receptor by a mechanism analogous to tachyphylaxis. Sartans/bisartans were found to bind to alpha adrenergic receptors resulting in structure-dependent desensitization or resensitization. These considerations have provided information on the mechanisms of receptor desensitization/tolerance and insights into possible avenues for treating addiction. In this regard sartans, which appear to cross the blood-brain barrier more readily than bisartans, are the preferred drug candidates.
Collapse
Affiliation(s)
- Graham J. Moore
- Pepmetics Inc., 772 Murphy Place, Victoria, BC V8Y 3H4, Canada;
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Harry Ridway
- Institute for Sustainable Industries and Liveable Cities, Victoria University, Melbourne, VIC 8001, Australia;
| | - Laura Kate Gadanec
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
- Immunology Program, Australian Institute for Musculoskeletal Science (AIMSS), Melbourne, VIC 3021, Australia
| | - Anthony Zulli
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
| | - Jordan Swiderski
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
| | | | - Veroniki P. Vidali
- Institute of Nanoscience and Nanotechnology, National Centre for Scientific Research “Demokritos”, 15341 Athens, Greece;
| | | | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece;
| | - John M. Matsoukas
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada
- Institute for Health and Sport, Immunology and Translational Research, Victoria University, Melbourne, VIC 3030, Australia; (L.K.G.); (V.A.); (A.Z.); (J.S.)
- NewDrug/NeoFar PC, Patras Science Park, 26504 Patras, Greece;
- Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
5
|
Ravani LV, Gewehr DM, Calomeni P, Gauza MDM, Pereira J, Cardoso R, Ribeiro HB, Bocchi E. Angiotensin Receptor-Neprilysin Inhibitor Effects on Atherosclerotic Cardiovascular Disease Events: A Meta-Analysis of Randomized Controlled Trials. Am J Cardiol 2023; 205:259-268. [PMID: 37619492 DOI: 10.1016/j.amjcard.2023.07.154] [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: 03/17/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 08/26/2023]
Abstract
Sacubitril-valsartan is an angiotensin receptor-neprilysin inhibitor (ARNI) associated with a decreased risk of death and hospitalization for selected patients with heart failure (HF). However, its association with improved atherosclerotic cardiovascular disease (ASCVD) events remains unclear. We performed a meta-analysis to evaluate the association of ARNI with ASCVD events in patients with HF. We systematically searched PubMed, Embase, Cochrane, and ClinicalTrials.gov for studies comparing ARNIs with angiotensin-converting enzyme inhibitors (ACEIs) or angiotensin receptor blockers (ARBs) in terms of myocardial infarction, stroke, angina pectoris, peripheral artery disease, and the composite end point in patients with HF. A total of 8 randomized controlled trials were included, with 17,541 patients assigned to either the ARNI (8,764 patients) or ACEi/ARB (8,777 patients) groups. The incidence of composite end point (risk ratio [RR] 1.03, 95% confidence interval [CI] 0.93 to 1.13, p = 0.63), myocardial infarction (RR 1.02, 95% CI 0.81 to 1.30, p = 0.85), angina pectoris (RR 0.96, 95% CI 0.80 to 1.17, p = 0.70), and stroke (RR 0.99, 95% CI 0.85 to 1.16, p = 0.93) were not statistically different between the ARNI and ACEi/ARB groups. However, ARNI was associated with a higher incidence of peripheral artery disease (RR 1.63, 95% CI 1.05 to 2.52, p = 0.03). In conclusion, this meta-analysis found no association between ARNI therapy and improved ASCVD events in patients with HF.
Collapse
Affiliation(s)
- Lis Victoria Ravani
- Department of Medicine, University of São Paulo Medical School, São Paulo, Brazil.
| | - Douglas Mesadri Gewehr
- Curitiba Heart Institute (INCOR Curitiba) and Denton Cooley Institute of Research Science and Technology,Curitiba, Paraná, Brazil
| | - Pedro Calomeni
- Department of Medicine, University of São Paulo Medical School, São Paulo, Brazil
| | - Mateus de Miranda Gauza
- Department of Medicine, Universidade da Região de Joinville, Joinville, Santa Catarina, Brazill
| | | | - Rhanderson Cardoso
- Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Henrique Barbosa Ribeiro
- Department of Medicine, Universidade da Região de Joinville, Joinville, Santa Catarina, Brazill; Instituto do Coração, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Edimar Bocchi
- Department of Medicine, Universidade da Região de Joinville, Joinville, Santa Catarina, Brazill; Instituto do Coração, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
6
|
Lestari YM, Tarawan VM, Achadiyani A, Radhiyanti PT, Ray HRD, Lesmana R, Goenawan H. Exercise intensities modulate ACE2/MasR/eNOS pathway in male Wistar rat's lung. Physiol Rep 2023; 11:e15803. [PMID: 37667409 PMCID: PMC10477189 DOI: 10.14814/phy2.15803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 09/06/2023] Open
Abstract
Specific exercise intensities could improve lung vascular function by increasing nitric oxide (NO). The ACE2/MasR/eNOS axis is one of the pathways facilitating NO synthesis. This study examines the effect of different intensities of aerobic training on the ACE2/MasR/eNOS axis and histology of lung muscular arteries. Male Wistar rats were used in this study and randomized into control and exercise groups receiving low-, moderate-, and high-intensity training. The training was conducted for 30 min daily, five times a week, for 8 weeks. We observed that different exercise intensities affect the ACE2/MasR/eNOS pathway differently. Compared to control, high-intensity aerobic exercise significantly increased ACE2, Mas receptor (MasR), and eNOS mRNA expressions (p < 0.01). Moderate-intensity exercise significantly increased MasR and eNOS mRNA expressions compared to the control (p < 0.05), and this intensity also increased ACE2 mRNA but not significantly. Low-intensity exercise increased ACE2, MasR, and eNOS mRNA expressions but not significantly. Low-, moderate-, or high-intensity exercises reduced the medial wall thickness of the lung muscular arteries but not significantly. In conclusion, high-intensity exercise may induce NO synthesis in the lung by increasing mRNA expression of ACE2, MasR, and eNOS without decreasing the medial wall thickness of the muscular artery. Thus, high-intensity exercise may be the optimal intensity to improve NO synthesis and vascular function in the lung.
Collapse
Affiliation(s)
- Yani Medina Lestari
- Biomedical Science Master Program, Faculty of MedicineUniversitas PadjadjaranBandungIndonesia
| | - Vita Murniati Tarawan
- Department of Biomedical Science, Faculty of MedicineUniversitas PadjadjaranJatinangorIndonesia
| | - Achadiyani Achadiyani
- Department of Biomedical Science, Faculty of MedicineUniversitas PadjadjaranJatinangorIndonesia
| | - Putri Teesa Radhiyanti
- Department of Biomedical Science, Faculty of MedicineUniversitas PadjadjaranJatinangorIndonesia
| | | | - Ronny Lesmana
- Department of Biomedical Science, Faculty of MedicineUniversitas PadjadjaranJatinangorIndonesia
- Central LaboratoryUniversitas PadjadjaranJatinangorIndonesia
| | - Hanna Goenawan
- Department of Biomedical Science, Faculty of MedicineUniversitas PadjadjaranJatinangorIndonesia
- Central LaboratoryUniversitas PadjadjaranJatinangorIndonesia
| |
Collapse
|
7
|
Mallén A, Rodriguez-Urquia R, Alvarez R, Dorca-Duch E, Navarro E, Hueso M. Sex Differences in Glomerular Lesions, in Atherosclerosis Progression, and in the Response to Angiotensin-Converting Enzyme Inhibitors in the ApoE -/- Mice Model. Int J Mol Sci 2023; 24:13442. [PMID: 37686247 PMCID: PMC10487579 DOI: 10.3390/ijms241713442] [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: 08/07/2023] [Revised: 08/21/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
This study analyzes sex-based differences in renal structure and the response to the Angiotensin-Converting Enzyme (ACE) inhibitor enalapril in a mouse model of atherosclerosis. Eight weeks old ApoE-/- mice received enalapril (5 mg/kg/day, subcutaneous) or PBS (control) for an additional 14 weeks. Each group consisted of six males and six females. Females exhibited elevated LDL-cholesterol levels, while males presented higher creatinine levels and proteinuria. Enalapril effectively reduced blood pressure in both groups, but proteinuria decreased significantly only in females. Plaque size analysis and assessment of kidney inflammation revealed no significant sex-based differences. However, males displayed more severe glomerular injury, with increased mesangial expansion, mesangiolysis, glomerular foam cells, and activated parietal epithelial cells (PECs). Enalapril mitigated mesangial expansion, glomerular inflammation (particularly in the female group), and hypertrophy of the PECs in males. This study demonstrates sex-based differences in the response to enalapril in a mouse model of atherosclerosis. Males exhibited more severe glomerular injury, while enalapril provided renal protection, particularly in females. These findings suggest potential sex-specific considerations for ACE inhibitor therapy in chronic kidney disease and atherosclerosis cardiovascular disease. Further research is needed to elucidate the underlying mechanism behind these observations.
Collapse
Affiliation(s)
- Adrián Mallén
- Experimental Nephrology Laboratory, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08907 L’Hospitalet de Llobregat, Spain; (A.M.); (E.N.)
| | - Ronny Rodriguez-Urquia
- Department of Nephrology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.R.-U.); (R.A.)
| | - Rafael Alvarez
- Department of Nephrology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.R.-U.); (R.A.)
| | - Eduard Dorca-Duch
- Department of Pathology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain;
| | - Estanis Navarro
- Experimental Nephrology Laboratory, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08907 L’Hospitalet de Llobregat, Spain; (A.M.); (E.N.)
| | - Miguel Hueso
- Experimental Nephrology Laboratory, Institut d’Investigació Biomèdica de Bellvitge-IDIBELL, 08907 L’Hospitalet de Llobregat, Spain; (A.M.); (E.N.)
- Department of Nephrology, Hospital Universitari Bellvitge, 08907 L’Hospitalet de Llobregat, Spain; (R.R.-U.); (R.A.)
| |
Collapse
|
8
|
Eshraghi-Jazi F, Nematbakhsh M. Age- and Gender-Related Differences in Renal Vascular Responses to Angiotensin II in Rats: The Role of the Mas Receptor. J Aging Res 2023; 2023:3560468. [PMID: 37622033 PMCID: PMC10447085 DOI: 10.1155/2023/3560468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/15/2023] [Accepted: 08/01/2023] [Indexed: 08/26/2023] Open
Abstract
Background Renal hemodynamic is influenced by both gender difference and age. Also, the Mas receptor (MasR) as one of the depressor components of the renin-angiotensin system which has more expression in females could postpone some dysfunctions associated with age, although the association between MasR and age in renal vascular responses to angiotensin II (Ang II) in male and female rats was well undefined. Therefore, the current study examined the effects of age and sex on systemic and renal vascular responses to graded doses of Ang II in Wistar rats with or without MasR antagonists (A779). Materials and Methods Anesthetized Wistar male and female rats with two age ranges of 8-12 and 24-28 weeks were exposed to cannulate venous and arterial vessels. After stability, mean arterial pressure (MAP), renal perfusion pressure (RPP), renal vascular resistance (RVR), and renal blood flow (RBF) were measured in response to the infusion of Ang II with or without A779. Results There were no significant differences in the base values of MAP, RPP, RBF, and RVR between the two genders in both the age ranges of 8-12 and 24-28 weeks. In addition, no significant gender difference was observed in the age ranges of the above mentioned parameters among the groups receiving vehicle or A779. Also, the infusion of vehicle or A779 could not significantly change the base values. On the other hand, the responses of RBF and RVR to Ang II revealed gender differences among 8-12-week groups (P < 0.05) but not in 24-28-week groups, while the blockade of MasR could not influence the responses in the age ranges. Conclusion It was concluded that age could impress sex difference in RBF and RVR responses to Ang II infusion and that MasR alone could not participate in these responses. In other words, MasR is not active under normal and acutely elevated Ang II levels.
Collapse
Affiliation(s)
- Fatemeh Eshraghi-Jazi
- Water & Electrolytes Research Center Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mehdi Nematbakhsh
- Water & Electrolytes Research Center Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
9
|
Luo JY, Du GL, Hao YM, Liu F, Zhang T, Fang BB, Li XM, Gao XM, Yang YN. AT1R gene rs389566 polymorphism contributes to MACCEs in hypertension patients. BMC Cardiovasc Disord 2023; 23:284. [PMID: 37270600 DOI: 10.1186/s12872-023-03223-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 04/02/2023] [Indexed: 06/05/2023] Open
Abstract
OBJECTIVE To investigate the possible association between AT1R gene polymorphisms and major adverse cardiovascular and cerebrovascular events (MACCEs) in hypertension patients combined with or without coronary artery disease (CAD) in Xinjiang. METHODS 374 CAD patients and 341 non-CAD individuals were enrolled as study participants and all of them have a hypertension diagnosis. AT1R gene polymorphisms were genotyped by SNPscan™ typing assays. During the follow-up in the clinic or by telephone interview, MACCEs were recorded. Kaplan-Meier curves and Cox survival analyses were used to explore the association between AT1R gene polymorphisms and the occurrence of MACCEs. RESULTS AT1R gene rs389566 was associated with MACCEs. The TT genotype of the AT1R gene rs389566 had a significantly higher probability of MACCEs than the AA + AT genotype (75.2% vs. 24.8%, P = 0.033). Older age (OR = 1.028, 95% CI: 1.009-1.0047, P = 0.003) and TT genotype of rs389566 (OR = 1.770, 95% CI: 1.148-2.729, P = 0.01) were risk factors of MACCEs. AT1R gene rs389566 TT genotype may be a predisposing factor for the occurrence of MACCEs in hypertensive patients. CONCLUSION We should also pay more attention to the prevent of MACCEs in hypertension patients combined with CAD. Especially those elderly hypertensive patients carrying AT1R rs389566 TT genotype requires avoidance of unhealthy lifestyle, better management of blood pressure control and reduce the occurrence of MACCEs.
Collapse
Affiliation(s)
- Jun-Yi Luo
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Guo-Li Du
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Yang-Min Hao
- Department of Endocrinology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Fen Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Tong Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Bin-Bin Fang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China
- Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiao-Mei Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China.
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China.
- Department of Cardiology, First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
- Xinjiang Key Laboratory of Medical Animal Model Research, Clinical Medical Research Institute of First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China.
| | - Yi-Ning Yang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, China.
- People's Hospital of Xinjiang Uygur Autonomous Region, 91 Tianchi Road, Urumqi, Xinjiang, China.
| |
Collapse
|
10
|
Shamsol Azman ANS, Tan JJ, Abdullah MNH, Bahari H, Lim V, Yong YK. Network Pharmacology and Molecular Docking Analysis of Active Compounds in Tualang Honey against Atherosclerosis. Foods 2023; 12:foods12091779. [PMID: 37174317 PMCID: PMC10178747 DOI: 10.3390/foods12091779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/12/2023] [Accepted: 04/17/2023] [Indexed: 05/15/2023] Open
Abstract
Atherosclerosis, a pathological condition marked by the accumulation of lipids and fibrous substances in the arterial walls, is a leading cause of heart failure and death. The present study aimed to utilize network pharmacology to assess the potential pharmacological effects of bioactive compounds in Tualang honey on atherosclerosis. This is significant as previous studies have indicated the cardioprotective effects of Tualang honey, yet a comprehensive evaluation using network pharmacology has yet to be conducted. The bioactive compounds in Tualang honey were screened and the potential gene targets for these compounds were predicted through Swiss Target Prediction and SuperPred databases. Atherosclerosis genes were retrieved from the OMIM, DisGeNet, and GeneCards databases. The interaction between these compounds and atherosclerosis genes was established through protein-protein interaction, gene ontology, and KEGG pathway analysis. The results of these analyses were then further confirmed through molecular docking studies using the AutoDock Tools software. The results revealed that 6 out of 103 compounds in Tualang honey met the screening criteria, with a total of 336 potential gene targets, 238 of which were shared with atherosclerosis. Further analysis showed that these active compounds had a good affinity with key targets and were associated with biological processes related to protein phosphorylation and inflammation as well as pathways related to lipid and atherosclerosis and other signaling pathways. In conclusion, the study provides insight into the potential pharmacological effects of Tualang honey bioactive compounds on atherosclerosis, supporting its use as a promising treatment for the disease.
Collapse
Affiliation(s)
- Ain Nabila Syahira Shamsol Azman
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Jun Jie Tan
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas 13200, Penang, Malaysia
| | - Muhammad Nazrul Hakim Abdullah
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Hasnah Bahari
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| | - Vuanghao Lim
- Advanced Medical and Dental Institute, Universiti Sains Malaysia, Bertam, Kepala Batas 13200, Penang, Malaysia
| | - Yoke Keong Yong
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia
| |
Collapse
|
11
|
Tufaro V, Serruys PW, Räber L, Bennett MR, Torii R, Gu SZ, Onuma Y, Mathur A, Baumbach A, Bourantas CV. Intravascular imaging assessment of pharmacotherapies targeting atherosclerosis: advantages and limitations in predicting their prognostic implications. Cardiovasc Res 2023; 119:121-135. [PMID: 35394014 DOI: 10.1093/cvr/cvac051] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 02/17/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Intravascular imaging has been often used over the recent years to examine the efficacy of emerging therapies targeting plaque evolution. Serial intravascular ultrasound, optical coherence tomography, or near-infrared spectroscopy-intravascular ultrasound studies have allowed us to evaluate the effects of different therapies on plaque burden and morphology, providing unique mechanistic insights about the mode of action of these treatments. Plaque burden reduction, a decrease in necrotic core component or macrophage accumulation-which has been associated with inflammation-and an increase in fibrous cap thickness over fibroatheromas have been used as surrogate endpoints to assess the value of several drugs in inhibiting plaque evolution and improving clinical outcomes. However, some reports have demonstrated weak associations between the effects of novel treatments on coronary atheroma and composition and their prognostic implications. This review examines the value of invasive imaging in assessing pharmacotherapies targeting atherosclerosis. It summarizes the findings of serial intravascular imaging studies assessing the effects of different drugs on atheroma burden and morphology and compares them with the results of large-scale trials evaluating their impact on clinical outcome. Furthermore, it highlights the limited efficacy of established intravascular imaging surrogate endpoints in predicting the prognostic value of these pharmacotherapies and introduces alternative imaging endpoints based on multimodality/hybrid intravascular imaging that may enable more accurate assessment of the athero-protective and prognostic effects of emerging therapies.
Collapse
Affiliation(s)
- Vincenzo Tufaro
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | | | - Lorenz Räber
- Department of Cardiology, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Ryo Torii
- Department of Mechanical Engineering, University College London, London, UK
| | - Sophie Zhaotao Gu
- Division of Cardiovascular Medicine, University of Cambridge, Cambridge, UK
| | - Yoshinobu Onuma
- Department of Cardiology, National University of Ireland, Galway, Ireland
| | - Anthony Mathur
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Andreas Baumbach
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Yale University School of Medicine, New Haven, CT, USA
| | - Christos Vasileios Bourantas
- Department of Cardiology, Barts Heart Centre, Barts Health NHS Trust, London, UK
- Centre for Cardiovascular Medicine and Devices, William Harvey Research Institute, Queen Mary University of London, London, UK
- Institute of Cardiovascular Sciences, University College London, London, UK
| |
Collapse
|
12
|
Déglise S, Bechelli C, Allagnat F. Vascular smooth muscle cells in intimal hyperplasia, an update. Front Physiol 2023; 13:1081881. [PMID: 36685215 PMCID: PMC9845604 DOI: 10.3389/fphys.2022.1081881] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 12/12/2022] [Indexed: 01/05/2023] Open
Abstract
Arterial occlusive disease is the leading cause of death in Western countries. Core contemporary therapies for this disease include angioplasties, stents, endarterectomies and bypass surgery. However, these treatments suffer from high failure rates due to re-occlusive vascular wall adaptations and restenosis. Restenosis following vascular surgery is largely due to intimal hyperplasia. Intimal hyperplasia develops in response to vessel injury, leading to inflammation, vascular smooth muscle cells dedifferentiation, migration, proliferation and secretion of extra-cellular matrix into the vessel's innermost layer or intima. In this review, we describe the current state of knowledge on the origin and mechanisms underlying the dysregulated proliferation of vascular smooth muscle cells in intimal hyperplasia, and we present the new avenues of research targeting VSMC phenotype and proliferation.
Collapse
Affiliation(s)
| | | | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
13
|
Geng YJ, Smolensky M, Sum-Ping O, Hermida R, Castriotta RJ. Circadian rhythms of risk factors and management in atherosclerotic and hypertensive vascular disease: Modern chronobiological perspectives of an ancient disease. Chronobiol Int 2023; 40:33-62. [PMID: 35758140 PMCID: PMC10355310 DOI: 10.1080/07420528.2022.2080557] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 12/13/2022]
Abstract
Atherosclerosis, a chronic inflammatory disease of the arteries that appears to have been as prevalent in ancient as in modern civilizations, is predisposing to life-threatening and life-ending cardiac and vascular complications, such as myocardial and cerebral infarctions. The pathogenesis of atherosclerosis involves intima plaque buildup caused by vascular endothelial dysfunction, cholesterol deposition, smooth muscle proliferation, inflammatory cell infiltration and connective tissue accumulation. Hypertension is an independent and controllable risk factor for atherosclerotic cardiovascular disease (CVD). Conversely, atherosclerosis hardens the arterial wall and raises arterial blood pressure. Many CVD patients experience both atherosclerosis and hypertension and are prescribed medications to concurrently mitigate the two disease conditions. A substantial number of publications document that many pathophysiological changes caused by atherosclerosis and hypertension occur in a manner dependent upon circadian clocks or clock gene products. This article reviews progress in the research of circadian regulation of vascular cell function, inflammation, hemostasis and atherothrombosis. In particular, it delineates the relationship of circadian organization with signal transduction and activation of the renin-angiotensin-aldosterone system as well as disturbance of the sleep/wake circadian rhythm, as exemplified by shift work, metabolic syndromes and obstructive sleep apnea (OSA), as promoters and mechanisms of atherogenesis and risk for non-fatal and fatal CVD outcomes. This article additionally updates advances in the clinical management of key biological processes of atherosclerosis to optimally achieve suppression of atherogenesis through chronotherapeutic control of atherogenic/hypertensive pathological sequelae.
Collapse
Affiliation(s)
- Yong-Jian Geng
- The Center for Cardiovascular Biology and Atherosclerosis Research, Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Michael Smolensky
- The Center for Cardiovascular Biology and Atherosclerosis Research, Division of Cardiovascular Medicine, Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Department of Biomedical Engineering, Cockrell School of Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Oliver Sum-Ping
- The Center for Sleep Sciences and Medicine, Department of Psychiatry and Behavioral Sciences, School of Medicine, Stanford University, Stanford, CA, USA
| | - Ramon Hermida
- Bioengineering & Chronobiology Laboratories, Atlantic Research Center for Telecommunication Technologies (atlanTTic), University of Vigo, Vigo, Spain
| | - Richard J. Castriotta
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, Keck Medical School, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
14
|
Jadli AS, Ballasy NN, Gomes KP, Mackay CDA, Meechem M, Wijesuriya TM, Belke D, Thompson J, Fedak PWM, Patel VB. Attenuation of Smooth Muscle Cell Phenotypic Switching by Angiotensin 1-7 Protects against Thoracic Aortic Aneurysm. Int J Mol Sci 2022; 23:ijms232415566. [PMID: 36555207 PMCID: PMC9779869 DOI: 10.3390/ijms232415566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/13/2022] Open
Abstract
Thoracic aortic aneurysm (TAA) involves extracellular matrix (ECM) remodeling of the aortic wall, leading to reduced biomechanical support with risk of aortic dissection and rupture. Activation of the renin-angiotensin system, and resultant angiotensin (Ang) II synthesis, is critically involved in the onset and progression of TAA. The current study investigated the effects of angiotensin (Ang) 1-7 on a murine model of TAA. Male 8-10-week-old ApoEKO mice were infused with Ang II (1.44 mg/kg/day) and treated with Ang 1-7 (0.576 mg/kg/day). ApoEKO mice developed advanced TAA in response to four weeks of Ang II infusion. Echocardiographic and histological analyses demonstrated increased aortic dilatation, excessive structural remodelling, perivascular fibrosis, and inflammation in the thoracic aorta. Ang 1-7 infusion led to attenuation of pathological phenotypic alterations associated with Ang II-induced TAA. Smooth muscle cells (SMCs) isolated from adult murine thoracic aorta exhibited excessive mitochondrial fission, oxidative stress, and hyperproliferation in response to Ang II. Treatment with Ang 1-7 resulted in inhibition of mitochondrial fragmentation, ROS generation, and hyperproliferation. Gene expression profiling used for characterization of the contractile and synthetic phenotypes of thoracic aortic SMCs revealed preservation of the contractile phenotype with Ang 1-7 treatment. In conclusion, Ang 1-7 prevented Ang II-induced vascular remodeling and the development of TAA. Enhancing Ang 1-7 actions may provide a novel therapeutic strategy to prevent or delay the progression of TAA.
Collapse
Affiliation(s)
- Anshul S. Jadli
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Noura N. Ballasy
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Karina P. Gomes
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Cameron D. A. Mackay
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Megan Meechem
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Tishani Methsala Wijesuriya
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Darrell Belke
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Jennifer Thompson
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Alberta Children’s Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Paul W. M. Fedak
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Section of Cardiac Surgery, Department of Cardiac Sciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Vaibhav B. Patel
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
- Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
- Correspondence: or ; Tel.: +1-(403)-220-3446
| |
Collapse
|
15
|
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
|
16
|
Gan Y, Feng Y, Zhou X, Li H, Wang G, Aini M, Shu J, Tu D. Serum levels of angiotensin-converting enzyme 2 in children with Kawasaki disease. Clin Exp Med 2022:10.1007/s10238-022-00933-x. [PMID: 36344782 PMCID: PMC10390598 DOI: 10.1007/s10238-022-00933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 10/25/2022] [Indexed: 11/09/2022]
Abstract
AbstractKawasaki disease (KD) has replaced rheumatic fever as the main cause of acquired heart disease in Japanese, American, and Chinese children. Polymorphisms in angiotensin-converting enzyme may be associated with susceptibility to KD, but the association of angiotensin-converting enzyme 2 (ACE2) with vascular endothelial injury in KD and the possibility for prognosis of vascular injury in KD by evaluating changes in serum ACE2 have not yet been assessed. Thus, this study aimed to investigate ACE2 levels in patients with KD to further explore the relationship between ACE2 and vascular injury in KD. Blood samples were collected from 49 children with KD before intravenous immunoglobulin treatment and 28 healthy children in the same period as the control group. Clinical data were collected from the patients and serum ACE2 levels of all participants were measured using an enzyme-linked immunosorbent assay. Serum ACE2 levels were significantly higher in the KD group than in the control group, and were negatively correlated with platelet levels in patients with KD. Serum ACE2 levels are related to the pathogenesis of KD and may be used as a potential serum marker for KD diagnosis.
Collapse
|
17
|
Novák J, Maceková S, Héžová R, Máchal J, Zlámal F, Hlinomaz O, Rezek M, Souček M, Vašků A, Slabý O, Bienertová-Vašků J. Polymorphism rs7079 in miR-31/-584 Binding Site in Angiotensinogen Gene Associates with Earlier Onset of Coronary Artery Disease in Central European Population. Genes (Basel) 2022; 13:1981. [PMID: 36360218 PMCID: PMC9690213 DOI: 10.3390/genes13111981] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 10/24/2022] [Accepted: 10/27/2022] [Indexed: 07/30/2023] Open
Abstract
Angiotensinogen (AGT) represents a key component of the renin-angiotensin-aldosterone system (RAAS). Polymorphisms in the 3' untranslated region (3'UTR) of the AGT gene may alter miRNA binding and cause disbalance in the RAAS. Within this study, we evaluated the possible association of AGT +11525C/A (rs7079) with the clinical characteristics of patients with coronary artery diseases (CAD). Selective coronarography was performed in 652 consecutive CAD patients. Clinical characteristics of the patients, together with peripheral blood samples for DNA isolation, were collected. The genotyping of rs7079 polymorphism was performed with TaqMan® SNP Genotyping Assays. We observed that patients with the CC genotype were referred for coronarography at a younger age compared to those with the AA+CA genotypes (CC vs. AA+CA: 59.1 ± 9.64 vs. 60.91 ± 9.5 (years), p = 0.045). Moreover, according to the logistic regression model, patients with the CC genotype presented more often with restenosis than those with the CA genotype (p = 0.0081). In conclusion, CC homozygotes for rs7079 present with CAD symptoms at a younger age compared with those with the AA+CA genotype, and they are more prone to present with restenosis compared with heterozygotes.
Collapse
Affiliation(s)
- Jan Novák
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
- Department of Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
- Second Department of Internal Medicine, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, 656 91 Brno, Czech Republic
- Ondrej Slaby Joint Research Group, Central European Institute of Technology and Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Soňa Maceková
- Second Department of Internal Medicine, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, 656 91 Brno, Czech Republic
| | - Renata Héžová
- Ondrej Slaby Joint Research Group, Central European Institute of Technology and Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Jan Máchal
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, 656 91 Brno, Czech Republic
- International Clinical Research Center, St Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Filip Zlámal
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Ota Hlinomaz
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, 656 91 Brno, Czech Republic
- International Clinical Research Center, St Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Michal Rezek
- Department of Cardiovascular Diseases, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, 656 91 Brno, Czech Republic
- International Clinical Research Center, St Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Miroslav Souček
- Second Department of Internal Medicine, St. Anne’s University Hospital and Faculty of Medicine, Masaryk University, 656 91 Brno, Czech Republic
- International Clinical Research Center, St Anne’s University Hospital, 656 91 Brno, Czech Republic
| | - Anna Vašků
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| | - Ondřej Slabý
- Ondrej Slaby Joint Research Group, Central European Institute of Technology and Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Julie Bienertová-Vašků
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, 625 00 Brno, Czech Republic
| |
Collapse
|
18
|
Plasma Angiotensin Converting Enzyme 2 (ACE2) Activity in Healthy Controls and Patients with Cardiovascular Risk Factors and/or Disease. J Pers Med 2022; 12:jpm12091495. [PMID: 36143280 PMCID: PMC9501250 DOI: 10.3390/jpm12091495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/05/2022] [Accepted: 09/08/2022] [Indexed: 11/18/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is an endogenous negative regulator of the renin-angiotensin system, a key factor in the development of cardiovascular disease (CVD). ACE2 is also used by SARS-CoV-2 for host cell entry. Given that COVID-19 is associated with hypercoagulability, it is timely to explore the potential relationship between plasma ACE2 activity and the coagulation profile. In this cross-sectional study, ACE2 activity and global coagulation assays (GCA) including thromboelastography, thrombin, and fibrin generation were measured in adult healthy controls (n = 123; mean age 41 ± 17 years; 35% male) and in patients with cardiovascular risk factors and/or disease (n = 258; mean age 65 ± 14 years; 55% male). ACE2 activity was significantly lower in controls compared to patients with cardiovascular risk factors and/or disease (median 0.10 (0.02, 3.33) vs. 5.99 (1.95, 10.37) pmol/mL/min, p < 0.001). Of the healthy controls, 48% had undetectable ACE2 activity. Controls with detectable ACE2 had lower maximum amplitude (p < 0.001). In patients with cardiovascular risk factors and/or disease, those in the 3rd tertile were older and male (p = 0.002), with a higher Framingham grade and increased number of cardiovascular risk factors (p < 0.001). In conclusion, plasma ACE2 activity is undetectable to very low in young healthy controls with minimal clinically relevant associations to GCA. Patients with cardiovascular risk factors and/or disease have increased plasma ACE2 activity, suggesting that it may be an important biomarker of endothelial dysfunction and atherosclerosis.
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Hypertension (HTN) is a well known risk factor for atherosclerosis and peripheral arterial disease (PAD). PAD affects more than 250 million people globally and is associated with worse clinical outcomes. Although multiple studies have been performed to evaluate treatment of HTN in patients with PAD, blood pressure management in this high-risk cohort remains poor. RECENT FINDINGS There has been conflicting evidence regarding blood pressure goals in PAD with some recent studies showing adverse outcomes with low blood pressure in this patient population. Current guidelines, however, continue to recommend treatment goals in PAD patients similar to patients without PAD. To date, no single antihypertensive drug class has shown a clear benefit in PAD population over other antihypertensive drug classes. SUMMARY Prospective randomized trials enrolling PAD patients are required that can shed light on optimum blood pressure target and also distinguish between different antihypertensive drugs in terms of reducing adverse outcomes.
Collapse
|
20
|
Metformin Reverses the Effects of Angiotensin 2 in Human Mammary Arteries by Modulating the Expression of Nitric Oxide Synthases. SERBIAN JOURNAL OF EXPERIMENTAL AND CLINICAL RESEARCH 2022. [DOI: 10.2478/sjecr-2022-0070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Angiotensin 2 impairs vascular function by activation of reactive oxygen species (ROS) production and development of endothelial dysfunction. Metformin, the first-line therapeutic agent for type 2 diabetes mellitus, has vascular protective properties, beyond its glucose lowering effects. The aim of the present study was to in-vestigate the interaction between metformin and angiotensin 2 in human internal mammary arteries harvested from patients with coronary heart disease undergoing revascularization procedure, by evaluation of vascular function, reactive oxygen species (ROS) production and the gene expression of nitric oxide (NO) synthases (endothelial – eNOS, neuronal – nNOS and inducible – iNOS). To this aim, vascular samples were incubated with angiotensin 2 (Ang2, 12 h) with/without metformin (Metf, 10 μM) and used for ROS measurement (FOX assay), vascular reactivity in organ bath (contractility to phenylephrine, relaxation to acetylcholine, con-tractility to NG-nitro-L-arginine methyl ester/L-NAME) and RT-PCT studies. Acute incubation of the vascular rings with Ang2 im-paired vascular reactivity (increase contractility, decrease relax-ation), increased ROS production, supressed eNOS/nNOS and in-creased iNOS mRNA expression. Ex vivo incubation with metfor-min at a clinically relevant concentration reversed all these ef-fects. These data suggest that Metformin might be useful in allevi-ating endothelial dysfunction by improving the endothelial-de-pendent relaxation and mitigating oxidative stress in clinical set-ting associated with cardiovascular disease regardless the pres-ence of impaired glucose metabolism.
Collapse
|
21
|
The Effect of Angiotensin Converting Enzyme (ACE) I/D Polymorphism on Atherosclerotic Cardiovascular Disease and Cardiovascular Mortality Risk in Non-Hemodialyzed Chronic Kidney Disease: The Mediating Role of Plasma ACE Level. Genes (Basel) 2022; 13:genes13071121. [PMID: 35885904 PMCID: PMC9318243 DOI: 10.3390/genes13071121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Revised: 06/18/2022] [Accepted: 06/20/2022] [Indexed: 02/05/2023] Open
Abstract
The association between angiotensin-converting enzyme insertion/deletion (ACE I/D) polymorphisms and plasma ACE levels may allow for the optimization of a preventive intervention to reduce cardiovascular morbidity and mortality in the chronic kidney disease (CKD) population. In this study, we aimed to analyze the association between ACE I/D polymorphism and cardiovascular mortality risk among non-hemodialyzed chronic kidney disease patients. This cross-sectional study examined 70 patients of Javanese ethnic origin with stable CKD who did not receive hemodialysis. ACE I/D polymorphisms, plasma ACE levels, atherosclerotic cardiovascular disease (ASCVD) risk, and cardiovascular mortality risk were investigated. As per our findings, the I allele was found to be more frequent (78.6) than the D allele (21.4), and the DD genotype was less frequent than the II genotype (4.3 vs. 61.4). The ACE I/D polymorphism had a significant direct positive effect on plasma ACE levels (path coefficient = 0.302, p = 0.021). Similarly, plasma ACE levels had a direct and significant positive effect on the risk of atherosclerotic cardiovascular disease (path coefficient = 0.410, p = 0.000). Moreover, atherosclerotic cardiovascular disease risk had a significant positive effect on cardiovascular mortality risk (path coefficient = 0.918, p = 0.000). The ACE I/D polymorphism had no direct effect on ASCVD and cardiovascular mortality risk. However, our findings show that the indirect effects of high plasma ACE levels may be a factor in the increased risk of ASCVD and cardiovascular mortality in Javanese CKD patients.
Collapse
|
22
|
Bian SZ, Zhang C, Rao RS, Ding XH, Huang L. Systemic Blood Predictors of Elevated Pulmonary Artery Pressure Assessed by Non-invasive Echocardiography After Acute Exposure to High Altitude: A Prospective Cohort Study. Front Cardiovasc Med 2022; 9:866093. [PMID: 35757324 PMCID: PMC9226344 DOI: 10.3389/fcvm.2022.866093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 05/06/2022] [Indexed: 11/14/2022] Open
Abstract
Aim Elevated pulmonary artery pressure (ePAP) in response to high-altitude hypoxia is a critical physiopathological factor in the hypoxic adaptation that may lead to high-altitude pulmonary edema in the acute phase or high-altitude pulmonary hypertension in the long term. However, the sea-level predictors of risk factors for altitude-induced ePAP have not been examined. Thus, we aimed to identify the baseline systemic blood predictors of ePAP after acute high-altitude exposure. Materials and Methods A total of 154 participants were transported to a high altitude 3,700 m from sea level within 2 h. Echocardiography examinations were performed to assess the mean pulmonary artery pressure (mPAP) and hemodynamics at both altitudes. All the individuals underwent blood tests to determine the concentrations of vascular regulatory factors. Univariate and adjusted logistic regression analyses were performed to identify the independent predictors of ePAP and factors related to ePAP. Results The mPAP increased significantly from sea level to high altitude (19.79 ± 6.53–27.16 ± 7.16 mmHg, p < 0.05). Increased levels of endothelin (ET-1), Ang (1–7), Ang II, and bradykinin were found after high-altitude exposure, while the levels of nitric oxide (NO), prostaglandin E2 (PEG2), and serotonin decreased sharply (all p-values < 0.05). At high altitude, 52.6% of the subjects exhibited ePAP, and the mPAP was closely correlated with the baseline Ang II level (r = 0.170, p = 0.036) and follow-up levels of NO (r = −0.209, p = 0.009), Ang II (r = 0.246, p = 0.002), and Ang (1–7) (r = −0.222, p = 0.006) and the left atrial inner diameter (LAD, r = 0.270, p < 0.001). Both the baseline and follow-up NO and Ang II levels were significantly different between the ePAP and non-ePAP groups. Finally, we identified the baseline Ang II and NO concentrations as two independent predictors of ePAP (p < 0.05). We also found that two vascular regulatory factors with inverse roles, namely, Ang (1–7) and Ang II, at high altitudes were independently associated with ePAP. Additionally, ET-1, NO, PEG2, and LAD were associated with ePAP. Conclusion The baseline concentrations of Ang II and NO at sea level are two independent predictors of ePAP after acute high-altitude exposure. Furthermore, Ang (1-7) and Ang II combined with ET-1, NO, PEG2, and LAD at high altitudes may contribute to the development of ePAP.
Collapse
Affiliation(s)
- Shi-Zhu Bian
- Department of Cardiology, Xinqiao Hospital, Institute of Cardiovascular Diseases, Army Medical University (Third Military Medical University), Chongqing, China
| | - Chen Zhang
- Department of Cardiology, Xinqiao Hospital, Institute of Cardiovascular Diseases, Army Medical University (Third Military Medical University), Chongqing, China
| | - Rong-Sheng Rao
- Department of Ultrasonography, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiao-Han Ding
- Department of Health Care and Geriatrics, The 940th Hospital of Joint Logistics Support of Chinese People’s Liberation Army (PLA), Lanzhou, China
| | - Lan Huang
- Department of Cardiology, Xinqiao Hospital, Institute of Cardiovascular Diseases, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Lan Huang,
| |
Collapse
|
23
|
Scisciola L, Fontanella RA, Surina, Garofalo G, Rizzo MR, Paolisso G, Barbieri M. Potential Role of Lisinopril in Reducing Atherosclerotic Risk: Evidence of an Antioxidant Effect in Human Cardiomyocytes Cell Line. Front Pharmacol 2022; 13:868365. [PMID: 35656292 PMCID: PMC9152216 DOI: 10.3389/fphar.2022.868365] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 03/21/2022] [Indexed: 11/17/2022] Open
Abstract
The cellular mechanisms involved in myocardial ischemia/reperfusion injury (I/R) pathogenesis are complex but attributable to reactive oxygen species (ROS) production. ROS produced by coronary endothelial cells, blood cells (e.g., leukocytes and platelets), and cardiac myocytes have the potential to damage vascular cells directly and cardiac myocytes, initiating mechanisms that induce apoptosis, inflammation, necrosis, and fibrosis of myocardial cells. In addition to reducing blood pressure, lisinopril, a new non-sulfhydryl angiotensin-converting enzyme (ACE) inhibitor, increases the antioxidant defense in animals and humans. Recently, it has been shown that lisinopril can attenuate renal oxidative injury in the l-NAME-induced hypertensive rat and cause an impressive improvement in the antioxidant defense system of Wistar rats treated with doxorubicin. The potential effect of lisinopril on oxidative damage and fibrosis in human cardiomyocytes has not been previously investigated. Thus, the present study aims to investigate the effect of different doses of lisinopril on oxidative stress and fibrotic mediators in AC16 human cardiomyocytes, along with a 7-day presence in the culture medium. The results revealed that AC16 human cardiomyocytes exposed to lisinopril treatment significantly showed an upregulation of proteins involved in protecting against oxidative stress, such as catalase, SOD2, and thioredoxin, and a reduction of osteopontin and Galectin-3, critical proteins involved in cardiac fibrosis. Moreover, lisinopril treatment induced an increment in Sirtuin 1 and Sirtuin 6 protein expression. These findings demonstrated that, in AC16 human cardiomyocytes, lisinopril could protect against oxidative stress and fibrosis via the activation of Sirtuin 1 and Sirtuin 6 pathways.
Collapse
Affiliation(s)
- Lucia Scisciola
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rosaria Anna Fontanella
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Surina
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giovanna Garofalo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Rosaria Rizzo
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Giuseppe Paolisso
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.,Mediterranea Cardiocentro, Napoli, Italy
| | - Michelangela Barbieri
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| |
Collapse
|
24
|
da Silva GM, da Silva MC, Nascimento DVG, Lima Silva EM, Gouvêa FFF, de França Lopes LG, Araújo AV, Ferraz Pereira KN, de Queiroz TM. Nitric Oxide as a Central Molecule in Hypertension: Focus on the Vasorelaxant Activity of New Nitric Oxide Donors. BIOLOGY 2021; 10:1041. [PMID: 34681140 PMCID: PMC8533285 DOI: 10.3390/biology10101041] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/08/2021] [Accepted: 10/09/2021] [Indexed: 12/15/2022]
Abstract
Cardiovascular diseases include all types of disorders related to the heart or blood vessels. High blood pressure is an important risk factor for cardiac complications and pathological disorders. An increase in circulating angiotensin-II is a potent stimulus for the expression of reactive oxygen species and pro-inflammatory cytokines that activate oxidative stress, perpetuating a deleterious effect in hypertension. Studies demonstrate the capacity of NO to prevent platelet or leukocyte activation and adhesion and inhibition of proliferation, as well as to modulate inflammatory or anti-inflammatory reactions and migration of vascular smooth muscle cells. However, in conditions of low availability of NO, such as during hypertension, these processes are impaired. Currently, there is great interest in the development of compounds capable of releasing NO in a modulated and stable way. Accordingly, compounds containing metal ions coupled to NO are being investigated and are widely recognized as having great relevance in the treatment of different diseases. Therefore, the exogenous administration of NO is an attractive and pharmacological alternative in the study and treatment of hypertension. The present review summarizes the role of nitric oxide in hypertension, focusing on the role of new NO donors, particularly the metal-based drugs and their protagonist activity in vascular function.
Collapse
Affiliation(s)
- Gabriela Maria da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Mirelly Cunha da Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Déborah Victória Gomes Nascimento
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Ellen Mayara Lima Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Fabíola Furtado Fialho Gouvêa
- School of Technical Health, Health Sciences Center, Federal University of Paraíba, João Pessoa 58.051-900, PB, Brazil;
| | - Luiz Gonzaga de França Lopes
- Laboratory of Bioinorganic Chemistry, Department of Organic and Inorganic Chemistry, Federal University of Ceará, Fortaleza 60.020-181, CE, Brazil;
| | - Alice Valença Araújo
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Kelli Nogueira Ferraz Pereira
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| | - Thyago Moreira de Queiroz
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, Vitória de Santo Antão 55.608-680, PE, Brazil; (G.M.d.S.); (M.C.d.S.); (D.V.G.N.); (E.M.L.S.); (A.V.A.); (K.N.F.P.)
| |
Collapse
|
25
|
The Level and Significance of Circulating Angiotensin-III in Patients with Coronary Atherosclerosis. J Renin Angiotensin Aldosterone Syst 2021; 2021:1704762. [PMID: 34603502 PMCID: PMC8478585 DOI: 10.1155/2021/1704762] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 08/08/2021] [Accepted: 08/14/2021] [Indexed: 01/17/2023] Open
Abstract
Objective Angiotensin-III (Ang-III) is the downstream product of angiotensin-II (Ang-II) metabolized by aminopeptidase A (APA). At present, the research of Ang-III mainly concentrates on hypertension and the central renin-angiotensin system (RAS). However, few studies have focused on the relationship between Ang-III and coronary atherosclerosis (CAS). Methods and Results Plasma Ang-III and APA levels were measured by the enzyme-linked immunosorbent assay (ELISA) in 44 normal subjects and 84 patients confirmed as having CAS by coronary angiography. Circulating Ang-III levels were significantly lower in patients with CAS than in normal controls (P = 0.013). APA levels were slightly lower in the CAS group (P = 0.324). According to the severity of atherosclerosis, CAS patients were divided into two groups. Compared with the controls, the APA and Ang-III levels were lower in the high scoring group and APA decreased significantly. Conclusions Circulating Ang-III levels were reduced in patients with CAS, and the possible reason may be related to the decrease in the APA level.
Collapse
|
26
|
Angiotensin-Converting Enzyme (ACE) 1 Gene Polymorphism and Phenotypic Expression of COVID-19 Symptoms. Genes (Basel) 2021; 12:genes12101572. [PMID: 34680966 PMCID: PMC8535484 DOI: 10.3390/genes12101572] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 12/25/2022] Open
Abstract
The renin–angiotensin–aldosterone system (RAAS) appears to play an important role in SARS-CoV-2 infection. Polymorphisms within the genes that control this enzymatic system are candidates for elucidating the pathogenesis of COVID-19, since COVID-19 is not only a pulmonary disease but also affects many organs and systems throughout the body in multiple ways. Most striking is the fact that ACE2, one of the major components of the RAAS, is a prerequisite for SARS-COV-2 infection. Recently, we and other groups reported an association between a polymorphism of the ACE1 gene (a homolog of ACE2) and the phenotypic expression of COVID-19, particularly in its severity. The ethnic difference in ACE1 insertion (I)/deletion (D) polymorphism seems to explain the apparent difference in mortality between the West and East Asia. The purpose of this review was to further evaluate the evidence linking ACE1 polymorphisms to COVID-19. We searched the Medline database (2019–2021) for reference citations of relevant articles and selected studies on the clinical outcome of COVID-19 related to ACE1 I/D polymorphism. Although the numbers of patients are not large enough yet, most available evidence supports the notion that the DD genotype adversely influences COVID-19 symptoms. Surprisingly, small studies conducted in several countries yielded opposite results, suggesting that the ACE1 II genotype is a risk factor. This contradictory result may be the case in certain geographic areas, especially in subgroups of patients. It may also be due to interactions with other genes or to yet unexplained biochemical mechanisms. According to our hypothesis, such candidates are genes that are functionally involved in the pathophysiology of COVID-19, can act in concert with the ACE1 DD genotype, and that show differences in their frequency between the West and East Asia. For this, we conducted research focusing on Alu-related genes. The current study on the ACE1 genotype will provide potentially new clues to the pathogenesis, treatment, and diagnosis of SARS-CoV-2 infections.
Collapse
|
27
|
Malekmohammad K, Bezsonov EE, Rafieian-Kopaei M. Role of Lipid Accumulation and Inflammation in Atherosclerosis: Focus on Molecular and Cellular Mechanisms. Front Cardiovasc Med 2021; 8:707529. [PMID: 34552965 PMCID: PMC8450356 DOI: 10.3389/fcvm.2021.707529] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis is a chronic lipid-driven and maladaptive inflammatory disease of arterial intima. It is characterized by the dysfunction of lipid homeostasis and signaling pathways that control the inflammation. This article reviews the role of inflammation and lipid accumulation, especially low-density lipoprotein (LDL), in the pathogenesis of atherosclerosis, with more emphasis on cellular mechanisms. Furthermore, this review will briefly highlight the role of medicinal plants, long non-coding RNA (lncRNA), and microRNAs in the pathophysiology, treatment, and prevention of atherosclerosis. Lipid homeostasis at various levels, including receptor-mediated uptake, synthesis, storage, metabolism, efflux, and its impairments are important for the development of atherosclerosis. The major source of cholesterol and lipid accumulation in the arterial wall is proatherogenic modified low-density lipoprotein (mLDL). Modified lipoproteins, such as oxidized low-density lipoprotein (ox-LDL) and LDL binding with proteoglycans of the extracellular matrix in the intima of blood vessels, cause aggregation of lipoprotein particles, endothelial damage, leukocyte recruitment, foam cell formation, and inflammation. Inflammation is the key contributor to atherosclerosis and participates in all phases of atherosclerosis. Also, several studies have shown that microRNAs and lncRNAs have appeared as key regulators of several physiological and pathophysiological processes in atherosclerosis, including regulation of HDL biogenesis, cholesterol efflux, lipid metabolism, regulating of smooth muscle proliferation, and controlling of inflammation. Thus, both lipid homeostasis and the inflammatory immune response are closely linked, and their cellular and molecular pathways interact with each other.
Collapse
Affiliation(s)
| | - Evgeny E. Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia
- Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Institute of Human Morphology, Moscow, Russia
- Institute for Atherosclerosis Research, Moscow, Russia
- Department of Biology and General Genetics, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Mahmoud Rafieian-Kopaei
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
28
|
Wang S, Tan Y, Yang T, Liu C, Li R. Pulmonary AngII promotes LPS-induced lung inflammation by regulating microRNA-143. Cytotechnology 2021; 73:745-754. [PMID: 34493899 PMCID: PMC8414951 DOI: 10.1007/s10616-021-00493-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 08/27/2021] [Indexed: 12/26/2022] Open
Abstract
Angiotensin converting enzyme 2 (ACE2) is a terminal carboxypeptidase, which cleaves single terminal residues from several bioactive peptides such as Angiotensin II (AngII). Many investigations indicated that ACE2 functions in angiotensin system and plays a crucial role in inflammatory lung diseases. However, the mechanism behind the involvement of ACE2 in inflammatory lung disease has not been fully elucidated. In this study, BEAS-2B cells were treated with gradient concentration of AngII and lipopolysaccharide (LPS) to induce inflammatory condition. Quantitative RT-PCR was performed to detect the level of ACE2 and miR-143-3p. Western blotting and immunofluorescence assays were performed to measure the expression of related proteins. The levels of inflammatory cytokines and cell viability were respectively measured by ELISA and CCK-8 kits. And ACE2 activity was detected by corresponding commercial kits. Bioinformatics methods were employed to predict the potential microRNA targeting ACE2, which was then confirmed by dual luciferase reporter assay. The results showed that ACE2 expression and activity were time-dependently decreased in LPS group for the first 12 h, after which this tendency was reversed. AngII addition enhanced these effects, compared with LPS group. Additionally, the lowest ACE2 activity level was found in both LPS and AngII + LPS groups at 6 h. The number of nuclei and the ACE2 expression were decreased in LPS groups at 6 h and further reduced by addition of AngII, detected by immunofluorescence. Moreover, ACE2 was validated to be a direct target of miR-143-3p. Pretreatment of AngII and LPS regulated the activity of ACE2, increased the expression of proinflammatory cytokines and cell apoptosis and regulated the expression of Bax, Bcl-2 and cleaved caspase-3 in BEAS-2B cells, which could be reversed by transfecting miR-143-3p inhibitor. The results collectively suggest that AngII promotes LPS-induced inflammation by regulating miR-143-3p in BEAS-2B cells. Therefore, miR-143-3p is considered a potential molecular target for the treatment of lung inflammation.
Collapse
Affiliation(s)
- Shenglan Wang
- Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China.,The Affiliated Hospital of Kunming University of Science and Technology, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China
| | - Yan Tan
- Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China.,The Affiliated Hospital of Kunming University of Science and Technology, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China
| | - Tingting Yang
- Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China.,The Affiliated Hospital of Kunming University of Science and Technology, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China
| | - Chen Liu
- Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China.,The Affiliated Hospital of Kunming University of Science and Technology, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China
| | - Rufang Li
- Pulmonary and Critical Care Medicine, The First People's Hospital of Yunnan Province, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China.,The Affiliated Hospital of Kunming University of Science and Technology, 157 Jinbi Road, Xishan District, Kunming, 650032 Yunnan China
| |
Collapse
|
29
|
Effects of Statins on Renin-Angiotensin System. J Cardiovasc Dev Dis 2021; 8:jcdd8070080. [PMID: 34357323 PMCID: PMC8305238 DOI: 10.3390/jcdd8070080] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Statins, a class of drugs for lowering serum LDL-cholesterol, have attracted attention because of their wide range of pleiotropic effects. An important but often neglected effect of statins is their role in the renin–angiotensin system (RAS) pathway. This pathway plays an integral role in the progression of several diseases including hypertension, heart failure, and renal disease. In this paper, the role of statins in the blockade of different components of this pathway and the underlying mechanisms are reviewed and new therapeutic possibilities of statins are suggested.
Collapse
|
30
|
Cajanding RJM. Comprehensive Review of Cardiovascular Involvement in COVID-19. AACN Adv Crit Care 2021; 32:169-187. [PMID: 33836538 DOI: 10.4037/aacnacc2021302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
COVID-19 has emerged as one of the most devastating and clinically significant infectious diseases of the last decade. It has reached global pandemic status at an unprecedented pace and has placed significant demands on health care systems worldwide. Although COVID-19 primarily affects the lungs, epidemiologic reports have shown that the disease affects other vital organs of the body, including the heart, vasculature, kidneys, brain, and the hematopoietic system. Of importance is the emerging awareness of the effects of COVID-19 on the cardiovascular system. The current state of knowledge regarding cardiac involvement in COVID-19 is presented in this article, with particular focus on the cardiovascular manifestations and complications of COVID-19 infection. The mechanistic insights of disease causation and the relevant pathophysiology involved in COVID-19 as they affect the heart are explored and described. Relevant practice essentials and clinical management implications for patients with COVID-19 with a cardiac pathology are presented in light of recent evidence.
Collapse
Affiliation(s)
- Ruff Joseph Macale Cajanding
- Ruff Joseph Macale Cajanding is Charge Nurse, Adult Critical Care Unit, 6th Floor, King George V Building, St Bartholomew's Hospital, Barts Health NHS Trust, 2 King Edward Street, London EC1A 1HQ, United Kingdom
| |
Collapse
|
31
|
Garcia-Garduño TC, Padilla-Gutierrez JR, Cambrón-Mora D, Valle Y. RAAS: A Convergent Player in Ischemic Heart Failure and Cancer. Int J Mol Sci 2021; 22:7106. [PMID: 34281199 PMCID: PMC8268500 DOI: 10.3390/ijms22137106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
The current global prevalence of heart failure is estimated at 64.34 million cases, and it is expected to increase in the coming years, especially in countries with a medium-low sociodemographic index where the prevalence of risk factors is increasing alarmingly. Heart failure is associated with many comorbidities and among them, cancer has stood out as a contributor of death in these patients. This connection points out new challenges both in the context of the pathophysiological mechanisms involved, as well as in the quality of life of affected individuals. A hallmark of heart failure is chronic activation of the renin-angiotensin-aldosterone system, especially marked by a systemic increase in levels of angiotensin-II, a peptide with pleiotropic activities. Drugs that target the renin-angiotensin-aldosterone system have shown promising results both in the prevention of secondary cardiovascular events in myocardial infarction and heart failure, including a lower risk of certain cancers in these patients, as well as in current cancer therapies; therefore, understanding the mechanisms involved in this complex relationship will provide tools for a better diagnosis and treatment and to improve the prognosis and quality of life of people suffering from these two deadly diseases.
Collapse
Affiliation(s)
- Texali C. Garcia-Garduño
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara 44340, Mexico; (T.C.G.-G.); (J.R.P.-G.)
- Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Jorge R. Padilla-Gutierrez
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara 44340, Mexico; (T.C.G.-G.); (J.R.P.-G.)
| | - Diego Cambrón-Mora
- Doctorado en Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Yeminia Valle
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara 44340, Mexico; (T.C.G.-G.); (J.R.P.-G.)
| |
Collapse
|
32
|
Poznyak AV, Bharadwaj D, Prasad G, Grechko AV, Sazonova MA, Orekhov AN. Renin-Angiotensin System in Pathogenesis of Atherosclerosis and Treatment of CVD. Int J Mol Sci 2021; 22:ijms22136702. [PMID: 34206708 PMCID: PMC8269397 DOI: 10.3390/ijms22136702] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/18/2021] [Accepted: 06/18/2021] [Indexed: 12/25/2022] Open
Abstract
Atherosclerosis has complex pathogenesis, which involves at least three serious aspects: inflammation, lipid metabolism alterations, and endothelial injury. There are no effective treatment options, as well as preventive measures for atherosclerosis. However, this disease has various severe complications, the most severe of which is cardiovascular disease (CVD). It is important to note, that CVD is among the leading causes of death worldwide. The renin–angiotensin–aldosterone system (RAAS) is an important part of inflammatory response regulation. This system contributes to the recruitment of inflammatory cells to the injured site and stimulates the production of various cytokines, such as IL-6, TNF-a, and COX-2. There is also an association between RAAS and oxidative stress, which is also an important player in atherogenesis. Angiotensin-II induces plaque formation at early stages, and this is one of the most crucial impacts on atherogenesis from the RAAS. Importantly, while stimulating the production of ROS, Angiotensin-II at the same time decreases the generation of NO. The endothelium is known as a major contributor to vascular function. Oxidative stress is the main trigger of endothelial dysfunction, and, once again, links RAAS to the pathogenesis of atherosclerosis. All these implications of RAAS in atherogenesis lead to an explicable conclusion that elements of RAAS can be promising targets for atherosclerosis treatment. In this review, we also summarize the data on treatment approaches involving cytokine targeting in CVD, which can contribute to a better understanding of atherogenesis and even its prevention.
Collapse
Affiliation(s)
- Anastasia V. Poznyak
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| | - Dwaipayan Bharadwaj
- Academy of Scientific and Innovative Research, CSIR-Institute of Genomics and Integrative Biology Campus, New Delhi 110067, India;
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Gauri Prasad
- Systems Genomics Laboratory, School of Biotechnology, Jawaharlal Nehru University, New Delhi 110067, India;
| | - Andrey V. Grechko
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, 14-3 Solyanka Street, 109240 Moscow, Russia;
| | - Margarita A. Sazonova
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
| | - Alexander N. Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, 121609 Moscow, Russia
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia;
- Institute of Human Morphology, 3 Tsyurupa Street, 117418 Moscow, Russia
- Correspondence: (A.V.P.); (A.N.O.)
| |
Collapse
|
33
|
Miličić Stanić B, Maddox S, de Souza AMA, Wu X, Mehranfard D, Ji H, Speth RC, Sandberg K. Male bias in ACE2 basic science research: missed opportunity for discovery in the time of COVID-19. Am J Physiol Regul Integr Comp Physiol 2021; 320:R925-R937. [PMID: 33848207 PMCID: PMC8203415 DOI: 10.1152/ajpregu.00356.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 11/22/2022]
Abstract
Throughout the world, including the United States, men have worse outcomes from COVID-19 than women. SARS-CoV-2, the causative virus of the COVID-19 pandemic, uses angiotensin-converting enzyme 2 (ACE2) to gain cellular entry. ACE2 is a member of the renin-angiotensin system (RAS) and plays an important role in counteracting the harmful effects mediated by the angiotensin type 1 receptor. Therefore, we conducted Ovid MEDLINE and Embase database searches of basic science studies investigating the impact of the biological variable of sex on ACE2 expression and regulation from 2000, the year ACE2 was discovered, through December 31, 2020. Out of 2,131 publications, we identified 853 original research articles on ACE2 conducted in primary cells, tissues, and/or whole mammals excluding humans. The majority (68.7%) of these studies that cited the sex of the animal were conducted in males, while 11.2% were conducted solely in females; 9.26% compared ACE2 between the sexes, while 10.8% did not report the sex of the animals used. General findings are that sex differences are tissue-specific and when present, are dependent upon gonadal state. Renal, cardiac, and adipose ACE2 is increased in both sexes under experimental conditions that model co-morbidities associated with worse COVID-19 outcomes including hypertension, obesity, and renal and cardiovascular diseases; however, ACE2 protein was generally higher in the males. Studies in Ace2 knockout mice indicate ACE2 plays a greater role in protecting the female from developing hypertension than the male. Studying the biological variable of sex in ACE2 research provides an opportunity for discovery in conditions involving RAS dysfunction and will shed light on sex differences in COVID-19 severity.
Collapse
Affiliation(s)
- Branka Miličić Stanić
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Sydney Maddox
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Aline M A de Souza
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Xie Wu
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Danial Mehranfard
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida
| | - Hong Ji
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| | - Robert C Speth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida
- Department of Pharmacology and Physiology, School of Medicine, Georgetown University, Washington, District of Columbia
| | - Kathryn Sandberg
- Center for the Study of Sex Differences in Health, Aging and Disease, Georgetown University, Washington, District of Columbia
| |
Collapse
|