1
|
Wang Z, Fan H, Wu J. Food-Derived Up-Regulators and Activators of Angiotensin Converting Enzyme 2: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:12896-12914. [PMID: 38810024 PMCID: PMC11181331 DOI: 10.1021/acs.jafc.4c01594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
Angiotensin-converting enzyme 2 (ACE2) is a key enzyme in the renin-angiotensin system (RAS), also serving as an amino acid transporter and a receptor for certain coronaviruses. Its primary role is to protect the cardiovascular system via the ACE2/Ang (1-7)/MasR cascade. Given the critical roles of ACE2 in regulating numerous physiological functions, molecules that can upregulate or activate ACE2 show vast therapeutic value. There are only a few ACE2 activators that have been reported, a wide range of molecules, including food-derived compounds, have been reported as ACE2 up-regulators. Effective doses of bioactive peptides range from 10 to 50 mg/kg body weight (BW)/day when orally administered for 1 to 7 weeks. Protein hydrolysates require higher doses at 1000 mg/kg BW/day for 20 days. Phytochemicals and vitamins are effective at doses typically ranging from 10 to 200 mg/kg BW/day for 3 days to 6 months, while Traditional Chinese Medicine requires doses of 1.25 to 12.96 g/kg BW/day for 4 to 8 weeks. ACE2 activation is linked to its hinge-bending region, while upregulation involves various signaling pathways, transcription factors, and epigenetic modulators. Future studies are expected to explore novel roles of ACE2 activators or up-regulators in disease treatments and translate the discovery to bedside applications.
Collapse
Affiliation(s)
- Zihan Wang
- Department
of Agricultural, Food and Nutritional Science, 4-10 Ag/For Building, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
- Cardiovascular
Research Centre, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| | - Hongbing Fan
- Department
of Animal and Food Sciences, University
of Kentucky, Lexington, Kentucky 40546, United States
| | - Jianping Wu
- Department
of Agricultural, Food and Nutritional Science, 4-10 Ag/For Building, University of Alberta, Edmonton, Alberta T6G 2P5, Canada
- Cardiovascular
Research Centre, University of Alberta, Edmonton, Alberta T6G 2R7, Canada
| |
Collapse
|
2
|
Rao A, Bhat SA, Shibata T, Giani JF, Rader F, Bernstein KE, Khan Z. Diverse biological functions of the renin-angiotensin system. Med Res Rev 2024; 44:587-605. [PMID: 37947345 DOI: 10.1002/med.21996] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 08/30/2023] [Accepted: 10/29/2023] [Indexed: 11/12/2023]
Abstract
The renin-angiotensin system (RAS) has been widely known as a circulating endocrine system involved in the control of blood pressure. However, components of RAS have been found to be localized in rather unexpected sites in the body including the kidneys, brain, bone marrow, immune cells, and reproductive system. These discoveries have led to steady, growing evidence of the existence of independent tissue RAS specific to several parts of the body. It is important to understand how RAS regulates these systems for a variety of reasons: It gives a better overall picture of human physiology, helps to understand and mitigate the unintended consequences of RAS-inhibiting or activating drugs, and sets the stage for potential new therapies for a variety of ailments. This review fulfills the need for an updated overview of knowledge about local tissue RAS in several bodily systems, including their components, functions, and medical implications.
Collapse
Affiliation(s)
- Adithi Rao
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Molecular, Cell and Developmental Biology, University of California Los Angeles, Los Angeles, California, USA
| | - Shabir A Bhat
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tomohiro Shibata
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jorge F Giani
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Florian Rader
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kenneth E Bernstein
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Zakir Khan
- Department of Pathology and Laboratory Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
3
|
Emslander Q, Krey K, Hamad S, Maidl S, Oubraham L, Hesse J, Henrici A, Austen K, Mergner J, Grass V, Pichlmair A. MDM2 Influences ACE2 Stability and SARS-CoV-2 Uptake. Viruses 2023; 15:1763. [PMID: 37632105 PMCID: PMC10459000 DOI: 10.3390/v15081763] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 07/20/2023] [Accepted: 08/07/2023] [Indexed: 08/27/2023] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is the central entry receptor for SARS-CoV-2. However, surprisingly little is known about the effects of host regulators on ACE2 localization, expression, and the associated influence on SARS-CoV-2 infection. Here we identify that ACE2 expression levels are regulated by the E3 ligase MDM2 and that MDM2 levels indirectly influence infection with SARS-CoV-2. Genetic depletion of MDM2 elevated ACE2 expression levels, which strongly promoted infection with all SARS-CoV-2 isolates tested. SARS-CoV-2 spike-pseudotyped viruses and the uptake of non-replication-competent virus-like particles showed that MDM2 affects the viral uptake process. MDM2 ubiquitinates Lysine 788 of ACE2 to induce proteasomal degradation, and degradation of this residue led to higher ACE2 expression levels and superior virus particle uptake. Our study illustrates that cellular regulators of ACE2 stability, such as MDM2, play an important role in defining the infection capabilities of SARS-CoV-2.
Collapse
Affiliation(s)
- Quirin Emslander
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Karsten Krey
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Sabri Hamad
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Susanne Maidl
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Lila Oubraham
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Joshua Hesse
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Alexander Henrici
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Katharina Austen
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Julia Mergner
- BayBioMS@MRI—Bavarian Center for Biomolecular Mass Spectrometry at Klinikum Rechts der Isar, Technical University of Munich, 81675 Munich, Germany
| | - Vincent Grass
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
| | - Andreas Pichlmair
- Institute of Virology, School of Medicine, Technical University of Munich (TUM), 81675 Munich, Germany (S.H.)
- German Centre for Infection Research (DZIF), Partner site Munich, 81675 Munich, Germany
- Center of Immunology of Viral Infection (CiViA), Aarhus University, 8000 Aarhus, Denmark
| |
Collapse
|
4
|
Furtado A, Esgalhado AJ, Duarte AC, Costa AR, Costa-Brito AR, Carro E, Ishikawa H, Schroten H, Schwerk C, Gonçalves I, Arosa FA, Santos CRA, Quintela T. Circadian rhythmicity of amyloid-beta-related molecules is disrupted in the choroid plexus of a female Alzheimer's disease mouse model. J Neurosci Res 2023; 101:524-540. [PMID: 36583371 DOI: 10.1002/jnr.25164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 11/20/2022] [Accepted: 12/18/2022] [Indexed: 12/31/2022]
Abstract
The choroid plexus (CP) is part of the blood-cerebrospinal fluid barrier (BCSFB) and was recently described as an important component of the circadian clock system. It is the principal source of cerebrospinal fluid (CSF) and responsible for the synthesis and secretion of various neuroprotective peptides including those involved in amyloid-β (Aβ) transport/degradation, contributing to Aβ homeostasis. Inadequate Aβ metabolic clearance and transport across the BCSFB have been associated with circadian dysfunctions in Alzheimer's disease (AD) patients. To investigate whether AD pathology influences Aβ scavengers circadian expression, we collected CP at different time points from an AD mouse model (APP/PS1) (female and male animals, aged 6- and 12-months-old) and analyzed their mRNA expression by Real-time RT-PCR. Only angiotensin-converting enzyme (Ace) expression in 6-month-old female wild-type mice and transthyretin (Ttr) expression in 12-month-old female wild-type mice presented significant rhythmicity. The circadian rhythmicity of Ace and Ttr, prompt us to analyze the involvement of circadian rhythm in Aβ uptake. A human CP papilloma (HIBCPP) cell line was incubated with Aβ-488 and uptake was evaluated at different time points using flow cytometry. Aβ uptake displayed circadian rhythmicity. Our results suggest that AD might affect Aβ scavengers rhythmicity and that Aβ clearance is a rhythmic process possibly regulated by the rhythmic expression of Aβ scavengers.
Collapse
Affiliation(s)
- André Furtado
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - André J Esgalhado
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ana C Duarte
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,UDI-IPG- Unidade de Investigação para o Desenvolvimento do Interior, Instituto Politécnico da Guarda, Guarda, Portugal
| | - Ana R Costa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Ana R Costa-Brito
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Eva Carro
- Networked Biomedical Research Center in Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Group of Neurodegenerative Diseases, Hospital 12 de Octubre Research Institute (imas12), Madrid, Spain
| | - Hiroshi Ishikawa
- Laboratory of Clinical Regenerative Medicine, Department of Neurosurgery, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Horst Schroten
- Mannheim Medical Faculty, University of Heidelberg, Childrens Hospital, Mannheim, Germany
| | - Christian Schwerk
- Mannheim Medical Faculty, University of Heidelberg, Childrens Hospital, Mannheim, Germany
| | - Isabel Gonçalves
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Fernando A Arosa
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Cecília R A Santos
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal
| | - Telma Quintela
- CICS-UBI, Health Sciences Research Centre, University of Beira Interior, Covilhã, Portugal.,UDI-IPG- Unidade de Investigação para o Desenvolvimento do Interior, Instituto Politécnico da Guarda, Guarda, Portugal
| |
Collapse
|
5
|
Devaux CA, Lagier JC. Unraveling the Underlying Molecular Mechanism of 'Silent Hypoxia' in COVID-19 Patients Suggests a Central Role for Angiotensin II Modulation of the AT1R-Hypoxia-Inducible Factor Signaling Pathway. J Clin Med 2023; 12:jcm12062445. [PMID: 36983445 PMCID: PMC10056466 DOI: 10.3390/jcm12062445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
A few days after being infected with SARS-CoV-2, a fraction of people remain asymptomatic but suffer from a decrease in arterial oxygen saturation in the absence of apparent dyspnea. In light of our clinical investigation on the modulation of molecules belonging to the renin angiotensin system (RAS) in COVID-19 patients, we propose a model that explains 'silent hypoxia'. The RAS imbalance caused by SARS-CoV-2 results in an accumulation of angiotensin 2 (Ang II), which activates the angiotensin 2 type 1 receptor (AT1R) and triggers a harmful cascade of intracellular signals leading to the nuclear translocation of the hypoxia-inducible factor (HIF)-1α. HIF-1α transactivates many genes including the angiotensin-converting enzyme 1 (ACE1), while at the same time, ACE2 is downregulated. A growing number of cells is maintained in a hypoxic condition that is self-sustained by the presence of the virus and the ACE1/ACE2 ratio imbalance. This is associated with a progressive worsening of the patient's biological parameters including decreased oxygen saturation, without further clinical manifestations. When too many cells activate the Ang II-AT1R-HIF-1α axis, there is a 'hypoxic spillover', which marks the tipping point between 'silent' and symptomatic hypoxia in the patient. Immediate ventilation is required to prevent the 'hypoxic spillover'.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
- Centre National de la Recherche Scientifique, 13000 Marseille, France
| | - Jean-Christophe Lagier
- Institut de Recherche pour le Développement, Assistance Publique Hôpitaux de Marseille, Microbes Evolution Phylogeny and Infection Laboratory, Aix-Marseille University, 13000 Marseille, France
- Institut Hospitalo-Universitaire-Méditerranée Infection, 13000 Marseille, France
| |
Collapse
|
6
|
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
|
7
|
Devaux CA, Raoult D. The impact of COVID-19 on populations living at high altitude: Role of hypoxia-inducible factors (HIFs) signaling pathway in SARS-CoV-2 infection and replication. Front Physiol 2022; 13:960308. [PMID: 36091390 PMCID: PMC9454615 DOI: 10.3389/fphys.2022.960308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Cases of coronavirus disease 2019 (COVID-19) have been reported worldwide. However, one epidemiological report has claimed a lower incidence of the disease in people living at high altitude (>2,500 m), proposing the hypothesis that adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection. This publication was initially greeted with skepticism, because social, genetic, or environmental parametric variables could underlie a difference in susceptibility to the virus for people living in chronic hypobaric hypoxia atmospheres. Moreover, in some patients positive for SARS-CoV-2, early post-infection ‘happy hypoxia” requires immediate ventilation, since it is associated with poor clinical outcome. If, however, we accept to consider the hypothesis according to which the adaptation to hypoxia may prove to be advantageous with respect to SARS-CoV-2 infection, identification of the molecular rational behind it is needed. Among several possibilities, HIF-1 regulation appears to be a molecular hub from which different signaling pathways linking hypoxia and COVID-19 are controlled. Interestingly, HIF-1α was reported to inhibit the infection of lung cells by SARS-CoV-2 by reducing ACE2 viral receptor expression. Moreover, an association of the rs11549465 variant of HIF-1α with COVID-19 susceptibility was recently discovered. Here, we review the evidence for a link between HIF-1α, ACE2 and AT1R expression, and the incidence/severity of COVID-19. We highlight the central role played by the HIF-1α signaling pathway in the pathophysiology of COVID-19.
Collapse
Affiliation(s)
- Christian Albert Devaux
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique, Marseille, France
- *Correspondence: Christian Albert Devaux,
| | - Didier Raoult
- Aix-Marseille University, IRD, APHM, MEPHI, Marseille, France
- IHU-Méditerranée Infection, Marseille, France
| |
Collapse
|
8
|
Stress-induced cardiometabolic perturbations, increased oxidative stress and ACE/ACE2 imbalance are improved by endurance training in rats. Life Sci 2022; 305:120758. [DOI: 10.1016/j.lfs.2022.120758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 11/22/2022]
|
9
|
Sengupta P, Dutta S. N-acetyl cysteine as a potential regulator of SARS-CoV-2-induced male reproductive disruptions. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2022; 27:14. [PMID: 35730047 PMCID: PMC9197722 DOI: 10.1186/s43043-022-00104-8] [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: 01/22/2022] [Accepted: 05/22/2022] [Indexed: 11/10/2022] Open
Abstract
Background The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), causing the coronavirus disease 2019 (COVID-19), has shown its persistent pandemic strength. This viral infectivity, kinetics, and the mechanisms of its actions in human body are still not completely understood. In addition, the infectivity and COVID-19 severity reportedly differ with patient’s gender with men being more susceptible to the disease. Thus, different studies have also suggested the adverse impact of COVID-19 on male reproductive functions, mainly emphasizing on high expressions of angiotensin-converting enzyme 2 (ACE2) in the testes that allows the viral entry into the cells. Main body The N-acetylcysteine (NAC), a potent therapeutic agent of COVID-19, may be effective in reducing the impairing impacts of this disease on male reproductive functions. NAC acts as mucolytic agent by reducing sulfide bonds in the cross-linked glycoprotein matrix in mucus owing to its free sulfhydryl group. Since NAC also breaks the viral disulfide bonds required for the host cell invasion, it may help to prevent direct SARS-CoV-2 invasion into the testicular cells as well. NAC also acts as a potent anti-inflammatory and antioxidant, directly scavenging reactive oxygen species (ROS) and regulating the redox state by maintaining the thiol pool being a precursor of cysteine (an essential substrate for glutathione synthesis). Since it is suggested that male reproductive impairment in COVID-19 patient may be caused by secondary immune responses owing to systemic inflammation and OS, the anti-inflammatory and antioxidant properties of NAC explained above may attribute in protecting the male reproduction functions from these COVID-19-mediated damages. Conclusion This article explains the mechanisms how NAC treatment for COVID-19 may prevent the infection-mediated disruptions in male reproduction.
Collapse
Affiliation(s)
- Pallav Sengupta
- Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jenjarom, Selangor Malaysia
| | - Sulagna Dutta
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Jenjarom, Selangor Malaysia
| |
Collapse
|
10
|
Cook JR, Ausiello J. Functional ACE2 deficiency leading to angiotensin imbalance in the pathophysiology of COVID-19. Rev Endocr Metab Disord 2022; 23:151-170. [PMID: 34195965 PMCID: PMC8245275 DOI: 10.1007/s11154-021-09663-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/04/2021] [Indexed: 02/07/2023]
Abstract
SARS-CoV-2, the virus responsible for COVID-19, uses angiotensin converting enzyme 2 (ACE2) as its primary cell-surface receptor. ACE2 is a key enzyme in the counter-regulatory pathway of the broader renin-angiotensin system (RAS) that has been implicated in a broad array of human pathology. The RAS is composed of two competing pathways that work in opposition to each other: the "conventional" arm involving angiotensin converting enzyme (ACE) generating angiotensin-2 and the more recently identified ACE2 pathway that generates angiotensin (1-7). Following the original SARS pandemic, additional studies suggested that coronaviral binding to ACE2 resulted in downregulation of the membrane-bound enzyme. Given the similarities between the two viruses, many have posited a similar process with SARS-CoV-2. Proponents of this ACE2 deficiency model argue that downregulation of ACE2 limits its enzymatic function, thereby skewing the delicate balance between the two competing arms of the RAS. In this review we critically examine this model. The available data remain incomplete but are consistent with the possibility that the broad multisystem dysfunction of COVID-19 is due in large part to functional ACE2 deficiency leading to angiotensin imbalance with consequent immune dysregulation and endothelial cell dysfunction.
Collapse
Affiliation(s)
- Joshua R Cook
- New York-Presbyterian Hospital and the Columbia University Irving Medical Center, New York, NY, USA
| | - John Ausiello
- New York-Presbyterian Hospital and the Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
11
|
Schettini IVG, Faria DV, Nogueira LS, Otoni A, Silva ACSE, Rios DRA. Renin angiotensin system molecules and chemokine (C-C motif) ligand 2 (CCL2) in chronic kidney disease patients. J Bras Nefrol 2022; 44:19-25. [PMID: 34251390 PMCID: PMC8943881 DOI: 10.1590/2175-8239-jbn-2021-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/26/2021] [Indexed: 11/22/2022] Open
Abstract
INTRODUCTION Studies have shown that the renin angiotensin aldosterone system (RAAS) and inflammation are related to kidney injury progression. The aim of this study was to evaluate RAAS molecules and chemokine (C-C motif) ligand 2 (CCL2) in 82 patients with chronic kidney disease (CKD). METHODS Patients were divided into two groups: patients diagnosed with CKD and patients without a CKD diagnosis. Glomerular filtration rate (GFR) and albumin/creatinine ratio (ACR) were determined, as well as plasma levels of angiotensin-(1-7) [Ang-(1-7)], angiotensin-converting enzyme (ACE)1, ACE2, and plasma and urinary levels of CCL2. RESULTS CCL2 plasma levels were significantly higher in patients with CKD compared to the control group. Patients with lower GFR had higher plasma levels of ACE2 and CCL2 and lower ratio ACE1/ACE2. Patients with higher ACR values had higher ACE1 plasma levels. CONCLUSION Patients with CKD showed greater activity of both RAAS axes, the classic and alternative, and higher plasma levels of CCL2. Therefore, plasma levels of RAAS molecules and CCL2 seem to be promising prognostic markers and even therapeutic targets for CKD.
Collapse
Affiliation(s)
| | - Débora Vargas Faria
- Universidade Federal de São João del-Rei, Campus Centro Oeste,
Divinópolis, MG, Brasil
| | | | - Alba Otoni
- Universidade Federal de São João del-Rei, Campus Centro Oeste,
Divinópolis, MG, Brasil
| | - Ana Cristina Simões e Silva
- Universidade Federal de Minas Gerais, Faculdade de Medicina,
Laboratório Interdisciplinar de Investigação Médica, Departamento de Pediatria, Belo
Horizonte, MG, Brasil
| | | |
Collapse
|
12
|
Faour WH, Choaib A, Issa E, Choueiry FE, Shbaklo K, Alhajj M, Sawaya RT, Harhous Z, Alefishat E, Nader M. Mechanisms of COVID-19-induced kidney injury and current pharmacotherapies. Inflamm Res 2022; 71:39-56. [PMID: 34802072 PMCID: PMC8606168 DOI: 10.1007/s00011-021-01520-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 10/31/2021] [Accepted: 11/01/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic created a worldwide debilitating health crisis with the entire humanity suffering from the deleterious effects associated with the high infectivity and mortality rates. While significant evidence is currently available online and targets various aspects of the disease, both inflammatory and noninflammatory kidney manifestations secondary to COVID-19 infection are still largely underrepresented. In this review, we summarized current knowledge about COVID-19-related kidney manifestations, their pathologic mechanisms as well as various pharmacotherapies used to treat patients with COVID-19. We also shed light on the effect of these medications on kidney functions that can further enhance renal damage secondary to the illness.
Collapse
Affiliation(s)
- Wissam H Faour
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon.
| | - Ali Choaib
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Elio Issa
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Francesca El Choueiry
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Khodor Shbaklo
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Maryline Alhajj
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Ramy Touma Sawaya
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Zeina Harhous
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, P.O. Box 36, Byblos, Lebanon
| | - Eman Alefishat
- Department of Pharmacology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi, United Arab Emirates
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, The University of Jordan, Amman, Jordan
| | - Moni Nader
- Department of Physiology and Immunology, College of Medicine and Health Sciences, Khalifa University of Science and Technology, P.O. Box 127788, Abu Dhabi, United Arab Emirates.
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, United Arab Emirates.
| |
Collapse
|
13
|
Hypertension May Reduce the Infection Risk but Increase the Severity of COVID-19: Based on the Current Data in China. Int J Hypertens 2021; 2021:6594863. [PMID: 34938578 PMCID: PMC8685758 DOI: 10.1155/2021/6594863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 11/03/2021] [Indexed: 12/15/2022] Open
Abstract
Increasing evidence has shown an unusual relationship between hypertension and COVID-19, which may not be as simple as previously thought. The purpose of our study was to determine the association of hypertension with the onset and development of COVID-19. A meta-analysis was performed to summarize the prevalence of hypertension in COVID-19 patients, as well as the usage of ACEIs/ARBs. Metaregression analyses were used to evaluate the association of hypertension with disease severity and mortality. PubMed and Google Scholar were searched for relevant studies. A total of 42 studies including 14138 patients were enrolled in the study. The proportion of hypertension in COVID-19 patients in China was 17.7% according to the enrolled studies, while it was 6.0% in a study containing 72314 confirmed cases, which are both much lower than in the general population. All of the data from the 11 provinces in China showed the same tendency. The proportions of hypertension were higher in severe/ICU patients and nonsurvivors than in nonsevere/ICU patients and survivors. The metaregression analyses suggested that both disease severity and risk of death were associated with the incidence of hypertension. A total of 27.6% of COVID-19 patients with hypertension received ACEI/ARB therapy. The proportion of deaths in COVID-19 patients with hypertension treated with ACEIs/ARBs was significantly lower than that in nonuse patients treated with ACEIs/ARBs. In conclusion, hypertension may reduce the infection risk of COVID-19 but increase the risk of developing worse clinical outcomes. The use of ACEIs/ARBs may benefit COVID-19 patients with hypertension.
Collapse
|
14
|
Chamata Y, Jackson KG, Watson KA, Jauregi P. Whey-Derived Peptides at the Heart of the COVID-19 Pandemic. Int J Mol Sci 2021; 22:11662. [PMID: 34769093 PMCID: PMC8584039 DOI: 10.3390/ijms222111662] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/14/2022] Open
Abstract
The renin-angiotensin system (RAS) is a key regulator of blood pressure and hypertension. Angiotensin-converting enzyme 2 (ACE2) and angiotensin-converting enzyme I (ACE) are two main components of the RAS that play a major role in blood pressure homeostasis. The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses ACE2 as a receptor to enter cells. Despite some controversies, numerous studies have reported a significant association between the use of ACE inhibitors and reduced risk of COVID-19. In our previous studies, we produced and identified peptide sequences present in whey hydrolysates exhibiting high ACE inhibitory activity. Therefore, the aim of this work is to obtain an improved understanding of the function of these natural peptides as RAS inhibitors and investigate their potential therapeutic role in the COVID-19 pandemic. The molecular interactions between peptides IPP, LIVTQ, IIAE, LVYPFP, and human ACE2 were assessed by employing a molecular docking approach. The results show that natural whey-derived peptides have a dual inhibitory action against both ACE and ACE2. This dual activity distinguishes these ACE inhibitory peptides from synthetic drugs, such as Captopril and Lisinopril which were not shown to inhibit ACE2 activity, and may represent a potential strategy in the treatment of COVID-19.
Collapse
Affiliation(s)
- Yara Chamata
- Harry Nursten Building, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (K.G.J.); (P.J.)
| | - Kim G. Jackson
- Harry Nursten Building, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (K.G.J.); (P.J.)
| | - Kimberly A. Watson
- Health and Life Sciences Building, School of Biological Sciences, University of Reading, Reading RG6 6EX, UK;
| | - Paula Jauregi
- Harry Nursten Building, Department of Food and Nutritional Sciences, University of Reading, Reading RG6 6DZ, UK; (K.G.J.); (P.J.)
| |
Collapse
|
15
|
Suleiman A, Rafaa T, Alrawi A, Dawood M. The impact of ACE2 genetic polymorphisms (rs2106809 and rs2074192) on gender susceptibility to COVID-19 infection and recovery: A systematic review. BAGHDAD JOURNAL OF BIOCHEMISTRY AND APPLIED BIOLOGICAL SCIENCES 2021. [DOI: 10.47419/bjbabs.v2i03.53] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: Epidemiological studies revealed there is a difference in susceptibility to infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) because of differences in gender with age and males being more inflicted. There is a clear indication that deaths caused by coronavirus disease 2019 (COVID-19) in males appeared at a higher rate than females across 35 nations. The implication of associated disease-risk genes, involved in the susceptibility of COVID-19 such as the angiotensin-converting enzyme 2 (ACE2), has recently received considerable attention due to their role in severe injury of lung and mediated SARS-CoV-2 entry as a host receptor.
Objectives: Herein, we aimed to systematically review how two main genetic polymorphisms of ACE2 (rs2106809 and rs2074192) can affect the gender susceptibility to SARS-CoV-2 infection.
Methods: To conduct this systematic review, a literature search in PubMed, Google Scholar, ScienceDirect, and Nature was made for the period 2004 to 2020. We searched for the impact of ACE2 genetic polymorphisms (rs2106809 and rs2074192) on gender susceptibility.
Results: We noticed that there was a differential genotype distribution between males and females in various global populations whereas mutant variants were common in males compared to wild-type variants among females, which may reflect differences in gender susceptibility to infection with SARS-CoV-2. Females are less susceptible to coronavirus as compare to males because of the expression of ACE2 receptor. It has a double role in favour of COVID-19 and against COVID-19.
Conclusions: Male mortality is greater than female mortality, which might be attributed to the ACE2 deficiency in women. Epidemiological studies have shown that the differences in sex and age have different susceptibility to SARS-CoV-2 infection.
Collapse
|
16
|
Zangaladze A, Cai CL, Marcelino M, Aranda JV, Beharry KD. Renal biomarkers of acute kidney injury in response to increasing intermittent hypoxia episodes in the neonatal rat. BMC Nephrol 2021; 22:299. [PMID: 34481475 PMCID: PMC8418040 DOI: 10.1186/s12882-021-02507-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 08/19/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND We tested the hypotheses that: 1) early exposure to increasing episodes of clinically relevant intermittent hypoxia (IH) is detrimental to the developing kidneys; and 2) there is a critical number of daily IH episodes which will result in irreparable renal damage that may involve angiotensin (Ang) II and endothelin (ET)-1. METHODS At birth (P0), neonatal rat pups were exposed to brief IH episodes from the first day of life (P0) to P7 or from P0-P14. Pups were either euthanized immediately or placed in room air (RA) until P21. RA littermates served as controls. Kidneys were harvested at P7, P14, and P21 for histopathology; angiotensin converting enzyme (ACE), ACE-2, ET-1, big ET-1, and malondialdehyde (MDA) levels; immunoreactivity of ACE, ACE-2, ET-1, ET-2, ET receptors (ETAR, ETBR), and hypoxia inducible factor (HIF)1α; and apoptosis (TUNEL stain). RESULTS Histopathology showed increased renal damage with 8-12 IH episodes/day, and was associated with Ang II, ACE, HIF1α, and apoptosis. ACE-2 was not expressed at P7, and minimally increased at P14. However, a robust ACE-2 response was seen during recovery with maximum levels noted in the groups recovering from 8 IH episodes/day. ET-1, big ET-1, ETAR, ETBR, and MDA increased with increasing levels of neonatal IH. CONCLUSIONS Chronic neonatal IH causes severe damage to the developing kidney with associated elevations in vasoconstrictors, suggesting hypertension, particularly with 8 neonatal IH episodes. ACE-2 is not activated in early postnatal life, and this may contribute to IH-induced vasoconstriction. Therapeutic targeting of ACE and ET-1 may help decrease the risk for kidney injury in the developing neonate to prevent and/or treat neonatal acute kidney injury and/or chronic kidney disease.
Collapse
Affiliation(s)
- Anano Zangaladze
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Charles L Cai
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Matthew Marcelino
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Jacob V Aranda
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
- Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
- SUNY Eye Institute, New York, NY, USA
| | - Kay D Beharry
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
- Department of Ophthalmology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA.
- SUNY Eye Institute, New York, NY, USA.
- Department of Pediatrics & Ophthalmology, Neonatal-Perinatal Medicine Clinical & Translational Research Labs, State University of New York, Downstate Medical Center, 450 Clarkson Avenue, Box 49, Brooklyn, NY, 11203, USA.
| |
Collapse
|
17
|
Sainsily X, Coquerel D, Giguère H, Dumont L, Tran K, Noll C, Ionescu AL, Côté J, Longpré JM, Carpentier A, Marsault É, Lesur O, Sarret P, Auger-Messier M. Elabela Protects Spontaneously Hypertensive Rats From Hypertension and Cardiorenal Dysfunctions Exacerbated by Dietary High-Salt Intake. Front Pharmacol 2021; 12:709467. [PMID: 34385922 PMCID: PMC8353398 DOI: 10.3389/fphar.2021.709467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/14/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives: Arterial hypertension, when exacerbated by excessive dietary salt intake, worsens the morbidity and mortality rates associated with cardiovascular and renal diseases. Stimulation of the apelinergic system appears to protect against several circulatory system diseases, but it remains unknown if such beneficial effects are conserved in severe hypertension. Therefore, we aimed at determining whether continuous infusion of apelinergic ligands (i.e., Apelin-13 and Elabela) exerted cardiorenal protective effects in spontaneously hypertensive (SHR) rats receiving high-salt diet. Methods: A combination of echocardiography, binding assay, histology, and biochemical approaches were used to investigate the cardiovascular and renal effects of Apelin-13 or Elabela infusion over 6 weeks in SHR fed with normal-salt or high-salt chow. Results: High-salt intake upregulated the cardiac and renal expression of APJ receptor in SHR. Importantly, Elabela was more effective than Apelin-13 in reducing high blood pressure, cardiovascular and renal dysfunctions, fibrosis and hypertrophy in high-salt fed SHR. Unlike Apelin-13, the beneficial effects of Elabela were associated with a counter-regulatory role of the ACE/ACE2/neprilysin axis of the renin-angiotensin-aldosterone system (RAAS) in heart and kidneys of salt-loaded SHR. Interestingly, Elabela also displayed higher affinity for APJ in the presence of high salt concentration and better resistance to RAAS enzymes known to cleave Apelin-13. Conclusion: These findings highlight the protective action of the apelinergic system against salt-induced severe hypertension and cardiorenal failure. As compared with Apelin-13, Elabela displays superior pharmacodynamic and pharmacokinetic properties that warrant further investigation of its therapeutic use in cardiovascular and kidney diseases.
Collapse
Affiliation(s)
- Xavier Sainsily
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - David Coquerel
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Hugo Giguère
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Lauralyne Dumont
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Kien Tran
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christophe Noll
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Andrei L Ionescu
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jérôme Côté
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Jean-Michel Longpré
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - André Carpentier
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Éric Marsault
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Olivier Lesur
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Philippe Sarret
- Département de Pharmacologie et Physiologie, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mannix Auger-Messier
- Département de Médecine, Centre de Recherche du CHUS, Faculté de Médecine et des Sciences de la Santé, Université de Sherbrooke, Sherbrooke, QC, Canada.,Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
18
|
Suganya S, Divya S, Parani M. Severe acute respiratory syndrome-coronavirus-2: Current advances in therapeutic targets and drug development. Rev Med Virol 2021; 31:e2174. [PMID: 32965078 PMCID: PMC7537282 DOI: 10.1002/rmv.2174] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/05/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022]
Abstract
The current pandemic of severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) has quickly emerged as a global health concern with government bodies worldwide taking drastic control measures. Understanding the virology of SARS-CoV-2, its molecular mechanisms, and its pathogenesis are required for a targeted therapeutic approach. In this review, we highlight the current molecular and drug advances that target SARS-CoV-2 at the genome level. We also summarize studies that therapeutically target the host angiotensin-converting enzyme 2 and proteases. Finally, we summarize antibody-mediated therapeutic approaches, as well as recent trends in vaccine development. Hence, the purpose of this study is to investigate different molecular targets in SARS-CoV-2 pathogenesis and their usefulness in developing strategies for drug development.
Collapse
Affiliation(s)
- Sakthivel Suganya
- Department of PsychologyTMC AcademySingapore
- Genome Research CenterAcademia SinicaTaipei CityTaiwan, ROC
| | - Suresh Divya
- Department of PediatricsNational Taiwan University HospitalYunlin CountyTaiwan, ROC
- Department of Genetic EngineeringGenomics LaboratorySRM Institute of Science and TechnologyKattankulathurTamil NaduIndia
| | - Madasamy Parani
- Department of Genetic EngineeringGenomics LaboratorySRM Institute of Science and TechnologyKattankulathurTamil NaduIndia
| |
Collapse
|
19
|
Siljee S, Milne B, Brasch HD, Bockett N, Patel J, Davis PF, Kennedy-Smith A, Itinteang T, Tan ST. Expression of Components of the Renin-Angiotensin System by Cancer Stem Cells in Renal Clear Cell Carcinoma. Biomolecules 2021; 11:537. [PMID: 33916968 PMCID: PMC8067590 DOI: 10.3390/biom11040537] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 12/23/2022] Open
Abstract
This study investigated the expression of components of the renin-angiotensin system (RAS) by cancer stem cells (CSCs) we have recently demonstrated in renal clear cell carcinoma (RCCC). Fifteen RCCC tissue samples underwent immunohistochemical staining for components of the RAS: renin, pro-renin receptor (PRR), angiotensin-converting enzyme (ACE), angiotensin-converting enzyme 2 (ACE2), and angiotensin II receptor 2 (AT2R). Immunofluorescence co-staining or double immunohistochemical staining of these components of the RAS with stemness-associated markers OCT4 or KLF4 was performed on two of the samples. Protein and transcript expression of these components of the RAS in six RCCC tissue samples was investigated using western blotting and reverse transcription quantitative polymerase chain reaction (RT-qPCR), respectively. In addition, angiotensin II receptor 1 (AT1R) was investigated using RT-qPCR only. Immunohistochemical staining demonstrated expression of renin, PRR, and ACE2 in 11, 13, and 13 out of 15 RCCC samples, respectively, while AT2R was expressed in all 15 samples. ACE was detected in the endothelium of normal vasculature only. Double immunohistochemical staining demonstrated localization of ACE2, but not renin, to the KLF4+ CSCs. Immunofluorescence staining showed localization of PRR and AT2R to the OCT4+ CSCs. Western blotting confirmed protein expression of all components of the RAS except renin. RT-qPCR demonstrated transcript expression of all components of the RAS including AT1R, but not AT2R, in all six RCCC tissue samples. This study demonstrated expression of PRR, ACE2, and AT2R by the CSCs within RCCC. Further studies may lead to novel therapeutic targeting of CSCs by manipulation of the RAS in the treatment of this aggressive cancer.
Collapse
Affiliation(s)
- Sam Siljee
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Bridget Milne
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Helen D. Brasch
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Nicholas Bockett
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Josie Patel
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Paul F. Davis
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Andrew Kennedy-Smith
- Department of Urology, Wellington Regional Hospital, Wellington 6021, New Zealand;
| | - Tinte Itinteang
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
| | - Swee T. Tan
- Gillies McIndoe Research Institute, Wellington 6242, New Zealand; (S.S.); (B.M.); (H.D.B.); (N.B.); (J.P.); (P.F.D.); (T.I.)
- Wellington Regional Plastic, Maxillofacial and Burns Unit, Hutt Hospital, Lower Hutt 5010, New Zealand
- Department of Surgery, The Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
20
|
Armaly Z, Kinaneh S, Skorecki K. Renal Manifestations of Covid-19: Physiology and Pathophysiology. J Clin Med 2021; 10:1216. [PMID: 33804075 PMCID: PMC8000200 DOI: 10.3390/jcm10061216] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/09/2021] [Accepted: 03/11/2021] [Indexed: 02/07/2023] Open
Abstract
Corona virus disease 2019 (COVID-19) imposes a serious public health pandemic affecting the whole world, as it is spreading exponentially. Besides its high infectivity, SARS-CoV-2 causes multiple serious derangements, where the most prominent is severe acute respiratory syndrome as well as multiple organ dysfunction including heart and kidney injury. While the deleterious impact of SARS-CoV-2 on pulmonary and cardiac systems have attracted remarkable attention, the adverse effects of this virus on the renal system is still underestimated. Kidney susceptibility to SARS-CoV-2 infection is determined by the presence of angiotensin-converting enzyme 2 (ACE2) receptor which is used as port of the viral entry into targeted cells, tissue tropism, pathogenicity and subsequent viral replication. The SARS-CoV-2 cellular entry receptor, ACE2, is widely expressed in proximal epithelial cells, vascular endothelial and smooth muscle cells and podocytes, where it supports kidney integrity and function via the enzymatic production of Angiotensin 1-7 (Ang 1-7), which exerts vasodilatory, anti-inflammatory, antifibrotic and diuretic/natriuretic actions via activation of the Mas receptor axis. Loss of this activity constitutes the potential basis for the renal damage that occurs in COVID-19 patients. Indeed, several studies in a small sample of COVID-19 patients revealed relatively high incidence of acute kidney injury (AKI) among them. Although SARS-CoV-1 -induced AKI was attributed to multiorgan failure and cytokine release syndrome, as the virus was not detectable in the renal tissue of infected patients, SARS-CoV-2 antigens were detected in kidney tubules, suggesting that SARS-CoV-2 infects the human kidney directly, and eventually induces AKI characterized with high morbidity and mortality. The mechanisms underlying this phenomenon are largely unknown. However, the fact that ACE2 plays a crucial role against renal injury, the deprivation of the kidney of this advantageous enzyme, along with local viral replication, probably plays a central role. The current review focuses on the critical role of ACE2 in renal physiology, its involvement in the development of kidney injury during SARS-CoV-2 infection, renal manifestations and therapeutic options. The latter includes exogenous administration of Ang (1-7) as an appealing option, given the high incidence of AKI in this ACE2-depleted disorder, and the benefits of ACE2/Ang1-7 including vasodilation, diuresis, natriuresis, attenuation of inflammation, oxidative stress, cell proliferation, apoptosis and coagulation.
Collapse
Affiliation(s)
- Zaher Armaly
- Department of Nephrology, Nazareth Hospital, EMMS, Nazareth 16100, Israel;
- The Bar-Ilan University Azrieli Faculty of Medicine, Safed 1311502, Israel;
| | - Safa Kinaneh
- Department of Nephrology, Nazareth Hospital, EMMS, Nazareth 16100, Israel;
| | - Karl Skorecki
- The Bar-Ilan University Azrieli Faculty of Medicine, Safed 1311502, Israel;
| |
Collapse
|
21
|
Angiotensin-(1-7)-A Potential Remedy for AKI: Insights Derived from the COVID-19 Pandemic. J Clin Med 2021; 10:jcm10061200. [PMID: 33805760 PMCID: PMC8001321 DOI: 10.3390/jcm10061200] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/02/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Membrane-bound angiotensin converting enzyme (ACE) 2 serves as a receptor for the Sars-CoV-2 spike protein, permitting viral attachment to target host cells. The COVID-19 pandemic brought into light ACE2, its principal product angiotensin (Ang) 1-7, and the G protein-coupled receptor for the heptapeptide (MasR), which together form a still under-recognized arm of the renin–angiotensin system (RAS). This axis counteracts vasoconstriction, inflammation and fibrosis, generated by the more familiar deleterious arm of RAS, including ACE, Ang II and the ang II type 1 receptor (AT1R). The COVID-19 disease is characterized by the depletion of ACE2 and Ang-(1-7), conceivably playing a central role in the devastating cytokine storm that characterizes this disorder. ACE2 repletion and the administration of Ang-(1-7) constitute the therapeutic options currently tested in the management of severe COVID-19 disease cases. Based on their beneficial effects, both ACE2 and Ang-(1-7) have also been suggested to slow the progression of experimental diabetic and hypertensive chronic kidney disease (CKD). Herein, we report a further step undertaken recently, utilizing this type of intervention in the management of evolving acute kidney injury (AKI), with the expectation of renal vasodilation and the attenuation of oxidative stress, inflammation, renal parenchymal damage and subsequent fibrosis. Most outcomes indicate that triggering the ACE2/Ang-(1-7)/MasR axis may be renoprotective in the setup of AKI. Yet, there is contradicting evidence that under certain conditions it may accelerate renal damage in CKD and AKI. The nature of these conflicting outcomes requires further elucidation.
Collapse
|
22
|
Coto E, Avanzas P, Gómez J. The Renin-Angiotensin-Aldosterone System and Coronavirus Disease 2019. Eur Cardiol 2021; 16:e07. [PMID: 33737961 PMCID: PMC7967817 DOI: 10.15420/ecr.2020.30] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Accepted: 10/10/2020] [Indexed: 12/21/2022] Open
Abstract
The renin-aldosterone-angiotensin system (RAAS) plays an important role in the pathogenesis of coronavirus disease 2019 (COVID-19), which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Angiotensin-converting enzyme 2 (ACE2) is the cellular receptor for SARS-CoV-2 and the host's expression of this membrane-bound protein could affect susceptibility to infection. The RAAS is an important regulator of cardiovascular physiology and ACE2 has an essential role. People with hypertension and other traits have shown to have an imbalance in ACE/ACE2 levels and reduced levels of ACE2 could enhance the risk of adverse outcome in patients with COVID-19. It has been hypothesised that the RAAS may mediate the interplay between cardiovascular disease and COVID-19 severity. Evidence shows that antihypertensive drugs that target the RAAS have no significant effect on the risk of infection and disease outcome. Variations in RAAS genes have been associated with the risk of developing hypertension and cardiovascular disease and could partly explain the heterogenous response to SARS-CoV-2 infection. This article explores the interplay between the RAAS and COVID-19, with emphasis on the possible relationship between genetic variations and disease severity.
Collapse
Affiliation(s)
- Eliecer Coto
- Genética Molecular, Hospital Universitario Central Asturias Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, ISPA Oviedo, Spain.,Universidad de Oviedo Oviedo, Spain.,Red de Investigación Renal (REDINREN) Madrid, Spain
| | - Pablo Avanzas
- Cardiología, Hospital Universitario Central Asturias Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, ISPA Oviedo, Spain.,Universidad de Oviedo Oviedo, Spain
| | - Juan Gómez
- Genética Molecular, Hospital Universitario Central Asturias Oviedo, Spain.,Instituto de Investigación Sanitaria del Principado de Asturias, ISPA Oviedo, Spain
| |
Collapse
|
23
|
Bhattacharyya N, Singh S, Halder A, Adhikari A, Ghosh R, Shikha D, Tripathi SK, Mallick AK, Mondal P, Pal SK. An Energy-Resolved Optical Non-invasive Device Detects Essential Electrolyte Balance in Humans at Point-of-Care. TRANSACTIONS OF THE INDIAN NATIONAL ACADEMY OF ENGINEERING : AN INTERNATIONAL JOURNAL OF ENGINEERING AND TECHNOLOGY 2021; 6:355-364. [PMID: 35837575 PMCID: PMC7878167 DOI: 10.1007/s41403-021-00204-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 01/21/2021] [Indexed: 06/15/2023]
Abstract
Regular monitoring of electrolyte balance is essential for patients suffering from chronic kidney disease (CKD), particularly those undergoing dialysis. In the context of the recent COVID-19 pandemic, more severe forms of infection are observed in elderly individuals and patients having co-morbidities like CKD. The repeated blood tests for the monitoring of electrolyte balance predispose them not only to COVID-19 but also other to hospital-acquired infections (HAI). Therefore, a non-invasive method for easy detection of essential electrolyte (K+ and Na+) levels is urgently needed. In this study, we developed an optical emission spectroscopy-based non-invasive device for simultaneous monitoring of salivary Na+ and K+ levels in a fast and reliable way. The device consisted of a closed spark chamber, micro-spectrometer, high voltage spark generator, electronic circuits, optical fiber, and an indigenously developed software based on the LabVIEW platform. The optical emission originating from the biological sample (i.e., saliva) due to recombination of ions energized by impingement of electrons returning from high voltage spark provides necessary information about the concentration of electrolytes. A small-scale clinical trial on 30 healthy human subjects shows the potential of the indigenously developed device in determining salivary Na + and K+ concentration. The low-cost, portable, point-of-care device requires only 2 mL of sample, and can simultaneously measure 1.0-190.0 mM Na+, and 1.0-270.9 mM K+ . To our understanding, the present work will find its relevance in combating COVID-19 morbidities, along with regular CKD patient-care.
Collapse
Affiliation(s)
- Neha Bhattacharyya
- Department of Radio Physics and Electronics, University of Calcutta, 92, Acharya Prafulla Chandra Rd, Machuabazar, Kolkata, 700009 India
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Soumendra Singh
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Animesh Halder
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
- Department of Applied Optics and Photonics, University of Calcutta, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Aniruddha Adhikari
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Ria Ghosh
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Deep Shikha
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| | - Santanu Kumar Tripathi
- Department of Clinical and Experimental Pharmacology, Calcutta School of Tropical Medicine, 108, Chittaranjan Avenue, Kolkata, 700073 India
| | - Asim Kumar Mallick
- Department of Paediatric Medicine, Nil Ratan Sircar Medical College and Hospital, 138, AJC Bose Road, Sealdah, Raja Bazar, Kolkata, 700014 India
| | - Pulak Mondal
- Department of Radio Physics and Electronics, University of Calcutta, 92, Acharya Prafulla Chandra Rd, Machuabazar, Kolkata, 700009 India
| | - Samir Kumar Pal
- Technical Research Centre, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
- Department of Chemical, Biological and Macromolecular Sciences, S. N. Bose National Centre for Basic Sciences, Block JD, Sector III, Salt Lake, Kolkata, 700106 India
| |
Collapse
|
24
|
Gao J, Liu Q, Zhao L, Yu J, Wang S, Cao T, Gao X, Wei Y. Identification and Antihypertension Study of Novel Angiotensin I-Converting Enzyme Inhibitory Peptides from the Skirt of Chlamys farreri Fermented with Bacillus natto. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:146-158. [PMID: 33356234 DOI: 10.1021/acs.jafc.0c04232] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The aim of this study was to isolate the angiotensin I-converting enzyme (ACE) inhibitory peptides from the skirt of Chlamys farreri fermented with Bacillus natto and to explore the antihypertension effect through in vivo studies. ACE inhibitory peptides were purified from the fermentation mixture by ultrafiltration, gel filtration chromatography, and reversed-phase high-performance liquid chromatography sequentially. The amino acids' sequence of the five novel ACE inhibitory peptides were identified by liquid chromatography-tandem mass spectrometry. Animal experiments demonstrated that the novel ACE inhibitory peptides significantly reduced the blood pressure in spontaneously hypertensive rats after a single or long-time treatment. Potential mechanisms were explored, and the results indicated that the novel peptides could regulate the renal renin-angiotensin system, improve vascular remodeling, inhibit myocardial fibrosis, and rebalance the gut microbial dysbiosis. Our results suggest that the fermentation products of the Chlamys farreri skirt by B. natto are potential sources of active peptides processing antihypertension activities.
Collapse
Affiliation(s)
- Jie Gao
- College of Life Sciences, Qingdao University, Qingdao 266071, China
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Qi Liu
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Ling Zhao
- Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China
| | - Jia Yu
- College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Shanglong Wang
- Chenland Nutritionals, Incorporated, Invine, California 92614, United States
| | - Tingfeng Cao
- Chenland Nutritionals, Incorporated, Invine, California 92614, United States
| | - Xiang Gao
- College of Life Sciences, Qingdao University, Qingdao 266071, China
| | - Yuxi Wei
- College of Life Sciences, Qingdao University, Qingdao 266071, China
| |
Collapse
|
25
|
Kianmehr A, Faraoni I, Kucuk O, Mahrooz A. Epigenetic alterations and genetic variations of angiotensin-converting enzyme 2 (ACE2) as a functional receptor for SARS-CoV-2: potential clinical implications. Eur J Clin Microbiol Infect Dis 2021; 40:1587-1598. [PMID: 33939044 PMCID: PMC8091148 DOI: 10.1007/s10096-021-04264-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
Receptor recognition is a crucial step in viral infection and is a critical factor for cell entry and tissue tropism. In several recent studies, angiotensin-converting enzyme 2 (ACE2) has been demonstrated to be the cellular receptor of SARS-CoV-2 as it was previously well known as the receptor of SARS-CoV. SARS-CoV-2 can bind with high affinity to human ACE2 and engages it as an entry receptor. It seems that the genetic, notably epigenetic variations of ACE2 are less known in different populations, indicating the need for its further investigation. These variations have the potential to affect its contribution to the pathogenicity of COVID-19. The contribution of epigenetics in the interindividual variability of ACE2 merits more attention because epigenetic processes can play important roles in ACE2 alterations in various tissues and different people and populations. Analyzing different DNA methylation patterns and microRNAs, contributing to the ACE2 modulation in the lungs will have a high priority. The epigenetic and genetic variations of ACE2 become even more important when considering that some people have mild clinical symptoms despite having COVID-19. The pathogenicity of SARS-CoV-2 infection is complex; therefore, a better understanding of the underlying pathobiology, especially binding the virus to its receptors, could help improve therapeutic and preventive approaches. This review aims to highlight the importance of evaluating both the epigenetic and genetic variations of ACE2 as a receptor for the deadly SARS-CoV-2.
Collapse
Affiliation(s)
- Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran ,Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Isabella Faraoni
- Department of Systems Medicine, University of Rome Tor Vergata, 00173 Rom, Italy
| | - Omer Kucuk
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA USA
| | - Abdolkarim Mahrooz
- Molecular and Cell Biology Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran ,Department of Clinical Biochemistry and Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Km 17 Khazarabad Road, Sari, Iran
| |
Collapse
|
26
|
Jiang X, Eales JM, Scannali D, Nazgiewicz A, Prestes P, Maier M, Denniff M, Xu X, Saluja S, Cano-Gamez E, Wystrychowski W, Szulinska M, Antczak A, Byars S, Skrypnik D, Glyda M, Król R, Zywiec J, Zukowska-Szczechowska E, Burrell LM, Woolf AS, Greenstein A, Bogdanski P, Keavney B, Morris AP, Heagerty A, Williams B, Harrap SB, Trynka G, Samani NJ, Guzik TJ, Charchar FJ, Tomaszewski M. Hypertension and renin-angiotensin system blockers are not associated with expression of angiotensin-converting enzyme 2 (ACE2) in the kidney. Eur Heart J 2020; 41:4580-4588. [PMID: 33206176 PMCID: PMC7665509 DOI: 10.1093/eurheartj/ehaa794] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 07/03/2020] [Accepted: 09/16/2020] [Indexed: 01/08/2023] Open
Abstract
AIMS Angiotensin-converting enzyme 2 (ACE2) is the cellular entry point for severe acute respiratory syndrome coronavirus (SARS-CoV-2)-the cause of coronavirus disease 2019 (COVID-19). However, the effect of renin-angiotensin system (RAS)-inhibition on ACE2 expression in human tissues of key relevance to blood pressure regulation and COVID-19 infection has not previously been reported. METHODS AND RESULTS We examined how hypertension, its major metabolic co-phenotypes, and antihypertensive medications relate to ACE2 renal expression using information from up to 436 patients whose kidney transcriptomes were characterized by RNA-sequencing. We further validated some of the key observations in other human tissues and/or a controlled experimental model. Our data reveal increasing expression of ACE2 with age in both human lungs and the kidney. We show no association between renal expression of ACE2 and either hypertension or common types of RAS inhibiting drugs. We demonstrate that renal abundance of ACE2 is positively associated with a biochemical index of kidney function and show a strong enrichment for genes responsible for kidney health and disease in ACE2 co-expression analysis. CONCLUSION Our results indicate that neither hypertension nor antihypertensive treatment is likely to alter the expression of the key entry receptor for SARS-CoV-2 in the human kidney. Our data further suggest that in the absence of SARS-CoV-2 infection, kidney ACE2 is most likely nephro-protective but the age-related increase in its expression within lungs and kidneys may be relevant to the risk of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xiao Jiang
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - James M Eales
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - David Scannali
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Alicja Nazgiewicz
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Priscilla Prestes
- School of Health and Life Sciences, Federation University Australia, Ballarat, VIC, Australia
| | - Michelle Maier
- School of Health and Life Sciences, Federation University Australia, Ballarat, VIC, Australia
| | - Matthew Denniff
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
| | - Xiaoguang Xu
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sushant Saluja
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Eddie Cano-Gamez
- Department of Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Wojciech Wystrychowski
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, Katowice, Poland
| | - Monika Szulinska
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Antczak
- Department of Urology and Uro-oncology, Karol Marcinkowski University of Medical Sciences, Poznan, Poland
| | - Sean Byars
- Centre for Systems Genomics, School of BioSciences, The University of Melbourne, Parkville, VIC, Australia
- Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Damian Skrypnik
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Maciej Glyda
- Department of Transplantology and General Surgery Poznan, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Robert Król
- Department of General, Vascular and Transplant Surgery, Medical University of Silesia, Katowice, Poland
| | - Joanna Zywiec
- Department of Internal Medicine, Diabetology and Nephrology, Medical University of Silesia, Zabrze, Poland
| | | | - Louise M Burrell
- Department of Medicine and Cardiology, University of Melbourne, Melbourne, VIC, Australia
| | - Adrian S Woolf
- Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Royal Manchester Children’s Hospital and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust, Manchester, UK
| | - Adam Greenstein
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Division of Medicine and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester, UK
| | - Pawel Bogdanski
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, Poznan University of Medical Sciences, Poznan, Poland
| | - Bernard Keavney
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Division of Medicine and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester, UK
| | - Andrew P Morris
- Division of Musculoskeletal & Dermatological Sciences, Faculty of Medicine, Biology and Health, University of Manchester, Manchester, UK
| | - Anthony Heagerty
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Division of Medicine and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester, UK
| | - Bryan Williams
- Institute of Cardiovascular Sciences, University College London, London, UK
| | - Stephen B Harrap
- Department of Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Gosia Trynka
- Department of Cellular Genetics, Wellcome Sanger Institute, Cambridge, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Leicester Biomedical Research Centre, National Institute for Health Research, Leicester, UK
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Department of Internal and Agricultural Medicine, Jagiellonian University College of Medicine, Kraków, Poland
| | - Fadi J Charchar
- School of Health and Life Sciences, Federation University Australia, Ballarat, VIC, Australia
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- Department of Physiology, University of Melbourne, Melbourne, VIC, Australia
| | - Maciej Tomaszewski
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
- Division of Medicine and Manchester Academic Health Science Centre, Manchester University NHS Foundation Trust Manchester, Manchester, UK
| |
Collapse
|
27
|
Kelleni MT. ACEIs, ARBs, ibuprofen originally linked to COVID-19: the other side of the mirror. Inflammopharmacology 2020; 28:1477-1480. [PMID: 32920716 PMCID: PMC7486973 DOI: 10.1007/s10787-020-00755-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/30/2020] [Indexed: 12/17/2022]
Abstract
During the COVID-19 pandemic, a correspondence, published at the Lancet Respiratory Medicine, that linked angiotensin-converting enzyme inhibitors, angiotensin receptor blockers and ibuprofen to a higher risk of SARS CoV-2 infection and complications, has influenced, when adopted by official health authorities, the practical management of COVID-19 with regard to non-steroidal anti-inflammatory drugs that were avoided in all COVID-19 management protocols all over the world. This manuscript discusses, from a pharmacological point of view, the points of weakness in the mentioned correspondence and it also lists some important contradictory review articles as well as clinical results that refuted its claims. The author chose to argue against each claim represented in the mentioned correspondence to confirm that ACEIs, ARBs and NSAIDs including ibuprofen should not be considered hazardous to be administered for COVID-19 patients and to warn against any future adoption of such unproved claims.
Collapse
Affiliation(s)
- Mina T Kelleni
- Pharmacology Department, College of Medicine, Minia University, Minya, Egypt.
| |
Collapse
|
28
|
Tavares CAM, Bailey MA, Girardi ACC. Biological Context Linking Hypertension and Higher Risk for COVID-19 Severity. Front Physiol 2020; 11:599729. [PMID: 33329052 PMCID: PMC7710931 DOI: 10.3389/fphys.2020.599729] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), represents a public health crisis of major proportions. Advanced age, male gender, and the presence of comorbidities have emerged as risk factors for severe illness or death from COVID-19 in observation studies. Hypertension is one of the most common comorbidities in patients with COVID-19. Indeed, hypertension has been shown to be associated with increased risk for mortality, acute respiratory distress syndrome, need for intensive care unit admission, and disease progression in COVID-19 patients. However, up to the present time, the precise mechanisms of how hypertension may lead to the more severe manifestations of disease in patients with COVID-19 remains unknown. This review aims to present the biological plausibility linking hypertension and higher risk for COVID-19 severity. Emphasis is given to the role of the renin-angiotensin system and its inhibitors, given the crucial role that this system plays in both viral transmissibility and the pathophysiology of arterial hypertension. We also describe the importance of the immune system, which is dysregulated in hypertension and SARS-CoV-2 infection, and the potential involvement of the multifunctional enzyme dipeptidyl peptidase 4 (DPP4), that, in addition to the angiotensin-converting enzyme 2 (ACE2), may contribute to the SARS-CoV-2 entrance into target cells. The role of hemodynamic changes in hypertension that might aggravate myocardial injury in the setting of COVID-19, including endothelial dysfunction, arterial stiffness, and left ventricle hypertrophy, are also discussed.
Collapse
Affiliation(s)
- Caio A M Tavares
- Geriatric Cardiology Unit, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Matthew A Bailey
- Centre for Cardiovascular Science, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana C C Girardi
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
29
|
Liang J, Lan J, Tang Q, Ling W, Li M. Short- and long-term treatment with angiotensin-converting enzyme inhibitors or calcium channel blockers for the prevention of diabetic nephropathy progression: A meta-analysis. Exp Ther Med 2020; 21:14. [PMID: 33235623 PMCID: PMC7678606 DOI: 10.3892/etm.2020.9446] [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: 05/21/2020] [Accepted: 10/14/2020] [Indexed: 11/05/2022] Open
Abstract
Treatments with angiotensin-converting enzyme (ACE) inhibitors or calcium channel blockers (CCBs) may delay the development of albuminuria in patients with early diabetic nephropathy. However, evidence in the literature has not been consistent. The present meta-analysis aimed to compare the short- and long-term therapeutic effects of ACE inhibitors and CCBs (when used separately) for preventing the progression of nephropathy in patients with diabetes mellitus. A comprehensive search of various databases was performed from inception until March 2015 for studies in the Chinese and English languages. Randomized controlled trials (RCTs) comparing the efficacy of ACE inhibitors with that of CCBs in patients with early diabetic nephropathy were considered. A total of 12 RCTs were included with a total of 947 patients. ACE inhibitors were indicated to be more effective in reducing the albumin excretion rate than CCBs after short-term treatments (<6 months) [mean difference (MD), 32.35; 95% confidence interval (CI), 31.62-33.07; P<0.00001]. There was no difference in serum creatinine values after treatment with either drug (MD, 8.7; 95% CI, -21.5-38.91; P=0.57). Data from six studies were used to compare long-term treatment effects (≥1 year). In terms of progression to normoalbuminuria, a marginal difference was obtained between the two drugs with better outcomes with ACE inhibitors [odds ratio (OR), 0.70; 95% CI, 0.49-1.00; P=0.05]. There was no statistically significant difference between ACE inhibitors and CCBs regarding the progression from microalbuminuria to macroalbuminuria (OR, 1.78; 95% CI, 0.82-3.87; P=0.15). In conclusion, the present study indicated that the antiproteinuric efficacy of CCBs may be less than that of ACE inhibitors after short-term treatment in patients with DN. However, both types of drugs are equally effective in reducing the progression of microalbuminuria to macroalbuminuria in the long term.
Collapse
Affiliation(s)
- Jialang Liang
- Endocrinology Department, Integrated Traditional Chinese and Western Medicine Hospital of Guangdong Province, Foshan, Guangdong 528200, P.R. China
| | - Jiarong Lan
- Nephrology Department, Huzhou Hospital of Traditional Chinese Medicine Affiliated Zhejiang University of Traditional Chinese Medicine, Huzhou, Zhejiang 313000, P.R. China
| | - Qizhi Tang
- Endocrinology Department, Integrated Traditional Chinese and Western Medicine Hospital of Guangdong Province, Foshan, Guangdong 528200, P.R. China
| | - Wenjing Ling
- Emergency Department, Integrated Traditional Chinese and Western Medicine Hospital of Guangdong Province, Foshan, Guangdong 528200, P.R. China
| | - Min Li
- Endocrinology Department, Huzhou Hospital of Traditional Chinese Medicine Affiliated Zhejiang University of Traditional Chinese Medicine, Huzhou, Zhejiang 313000, P.R. China
| |
Collapse
|
30
|
Gan R, Rosoman NP, Henshaw DJE, Noble EP, Georgius P, Sommerfeld N. COVID-19 as a viral functional ACE2 deficiency disorder with ACE2 related multi-organ disease. Med Hypotheses 2020; 144:110024. [PMID: 32758871 PMCID: PMC7308773 DOI: 10.1016/j.mehy.2020.110024] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 06/19/2020] [Indexed: 12/31/2022]
Abstract
SARS-CoV-2, the agent of COVID-19, shares a lineage with SARS-CoV-1, and a common fatal pulmonary profile but with striking differences in presentation, clinical course, and response to treatment. In contrast to SARS-CoV-1 (SARS), COVID-19 has presented as an often bi-phasic, multi-organ pathology, with a proclivity for severe disease in the elderly and those with hypertension, diabetes and cardiovascular disease. Whilst death is usually related to respiratory collapse, autopsy reveals multi-organ pathology. Chronic pulmonary disease is underrepresented in the group with severe COVID-19. A commonality of aberrant renin angiotensin system (RAS) is suggested in the at-risk group. The identification of angiotensin-converting-enzyme 2 (ACE2) as the receptor allowing viral entry to cells precipitated our interest in the role of ACE2 in COVID-19 pathogenesis. We propose that COVID-19 is a viral multisystem disease, with dominant vascular pathology, mediated by global reduction in ACE2 function, pronounced in disease conditions with RAS bias toward angiotensin-converting-enzyme (ACE) over ACE2. It is further complicated by organ specific pathology related to loss of ACE2 expressing cells particularly affecting the endothelium, alveolus, glomerulus and cardiac microvasculature. The possible upregulation in ACE2 receptor expression may predispose individuals with aberrant RAS status to higher viral load on infection and relatively more cell loss. Relative ACE2 deficiency leads to enhanced and protracted tissue, and vessel exposure to angiotensin II, characterised by vasoconstriction, enhanced thrombosis, cell proliferation and recruitment, increased tissue permeability, and cytokine production (including IL-6) resulting in inflammation. Additionally, there is a profound loss of the "protective" angiotensin (1-7), a vasodilator with anti-inflammatory, anti-thrombotic, antiproliferative, antifibrotic, anti-arrhythmic, and antioxidant activity. Our model predicts global vascular insult related to direct endothelial cell damage, vasoconstriction and thrombosis with a disease specific cytokine profile related to angiotensin II rather than "cytokine storm". Our proposed mechanism of lung injury provides an explanation for early hypoxia without reduction in lung compliance and suggests a need for revision of treatment protocols to address vasoconstriction, thromboprophylaxis, and to minimize additional small airways and alveolar trauma via ventilation choice. Our model predicts long term sequelae of scarring/fibrosis in vessels, lungs, renal and cardiac tissue with protracted illness in at-risk individuals. It is hoped that our model stimulates review of current diagnostic and therapeutic intervention protocols, particularly with respect to early anticoagulation, vasodilatation and revision of ventilatory support choices.
Collapse
Affiliation(s)
- Rosemary Gan
- Nambour Selangor Private Hospital, Nambour, QLD, Australia.
| | | | | | - Euan P Noble
- Sunshine Coast University Private Hospital, Birtinya, QLD, Australia
| | | | | |
Collapse
|
31
|
Khalaf K, Papp N, Chou JTT, Hana D, Mackiewicz A, Kaczmarek M. SARS-CoV-2: Pathogenesis, and Advancements in Diagnostics and Treatment. Front Immunol 2020; 11:570927. [PMID: 33123144 PMCID: PMC7573101 DOI: 10.3389/fimmu.2020.570927] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 09/04/2020] [Indexed: 12/15/2022] Open
Abstract
The emergence and rapid spread of SARS-CoV-2 in December 2019 has brought the world to a standstill. While less pathogenic than the 2002-2003 SARS-CoV, this novel betacoronavirus presents a global threat due to its high transmission rate, ability to invade multiple tissues, and ability to trigger immunological hyperactivation. The identification of the animal reservoir and intermediate host were important steps toward slowing the spread of disease, and its genetic similarity to SARS-CoV has helped to determine pathogenesis and direct treatment strategies. The exponential increase in cases has necessitated fast and reliable testing procedures. Although RT-PCR remains the gold standard, it is a time-consuming procedure, paving the way for newer techniques such as serologic tests and enzyme immunoassays. Various clinical trials using broad antiviral agents in addition to novel medications have produced controversial results; however, the advancement of immunotherapy, particularly monoclonal antibodies and immune modulators is showing great promise in clinical trials. Non-orthodox medications such as anti-malarials have been tested in multiple institutions but definitive conclusions are yet to be made. Adjuvant therapies have also proven to be effective in decreasing mortality in the disease course. While no formal guidelines have been established, the multitude of ongoing clinical trials as a result of unprecedented access to research data brings us closer to halting the SARS-CoV-2 pandemic.
Collapse
Affiliation(s)
- Khalil Khalaf
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Natalia Papp
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Jadzia Tin-Tsen Chou
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Doris Hana
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
| | - Andrzej Mackiewicz
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
| | - Mariusz Kaczmarek
- Department of Cancer Immunology, Poznan University of Medical Sciences, Poznań, Poland
- Department of Cancer Diagnostics and Immunology, Greater Poland Cancer Center, Poznań, Poland
| |
Collapse
|
32
|
Penna C, Mercurio V, Tocchetti CG, Pagliaro P. Sex-related differences in COVID-19 lethality. Br J Pharmacol 2020; 177:4375-4385. [PMID: 32698249 PMCID: PMC7405496 DOI: 10.1111/bph.15207] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/30/2020] [Accepted: 07/10/2020] [Indexed: 12/15/2022] Open
Abstract
Many countries have been affected by the worldwide outbreak of COVID-19. Among Western countries, Italy has been particularly hit at the beginning of the pandemic, immediately after China. In Italy and elsewhere, women seem to be less affected than men by severe/fatal COVID-19 infection, regardless of their age. Although women and men are affected differently by this infection, very few studies consider different therapeutic approaches for the two sexes. Understanding the mechanisms underlying these differences may help to find appropriate and sex specific therapies. Here, we consider that other mechanisms are involved to explain this difference, in addition to the protection attributable to oestrogens. Several X-linked genes (such as ACE2) and Y-linked genes (SRY and SOX9) may explain sex differences. Cardiovascular comorbidities are among the major enhancers of virus lethality. In addition, the number of sex-independent, non-genetic factors that can change susceptibility and mortality is enormous, and many other factors should be considered, including gender and cultural habits in different countries.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological SciencesUniversity of TorinoTurinItaly
- National Institute of Cardiovascular ResearchesBolognaItaly
| | - Valentina Mercurio
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
| | - Carlo G. Tocchetti
- Department of Translational Medical SciencesFederico II UniversityNaplesItaly
- Interdepartmental Center of Clinical and Translational ResearchFederico II UniversityNaplesItaly
| | - Pasquale Pagliaro
- Department of Clinical and Biological SciencesUniversity of TorinoTurinItaly
- National Institute of Cardiovascular ResearchesBolognaItaly
| |
Collapse
|
33
|
Ichikawa M, Konoshita T, Makino Y, Suzuki J, Ishizuka T, Nakamura H. An association study of C9orf3, a novel component of the renin-angiotensin system, and hypertension in diabetes. Sci Rep 2020; 10:16111. [PMID: 32999396 PMCID: PMC7528017 DOI: 10.1038/s41598-020-73094-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 07/21/2020] [Indexed: 12/14/2022] Open
Abstract
The renin-angiotensin system (RAS) is important in the onset and course of cardiovascular, kidney, and metabolic disorders. Previous reports showed that the RAS blockade protects organs and suppress the development of type 2 diabetes mellitus. A novel component of the RAS, namely, chromosome 9 open reading frame 3 (C9orf3), was recently identified, however, its effects are unclear. We evaluated whether the genetic variant of C9orf3 is associated with morbidity of hypertension among subjects with type 2 diabetes. We enrolled 382 subjects with type 2 diabetes, 222 of whom were diagnosed with hypertension. Human leukocyte genomic DNA was isolated and a genetic variant was analyzed for a C/T variant of C9orf3 (rs4385527) via PCR analysis. The relationship between the genotype and hypertension morbidity among subjects with diabetes was examined. The proportion of the respective C9orf3 genetic variants were as follows 247 CC, 119 CT, and 16 TT. The risk of hypertension was determined to be 1.58, with a 95% confidence interval of 1.11–2.27. Moreover, the p value was 0.012 for allelic comparison and for Armitage’s trend test, with the C allele identified as the risk factor. Consequently, hypertension was markedly associated with type 2 diabetes in subjects with the C9orf3 variant, exhibiting a nearly 1.6-fold increased risk. The C variant of a new component of the RAS, C9orf3 (rs4385527) might have a considerable impact on the pathogenesis of hypertension in diabetes.
Collapse
Affiliation(s)
- Mai Ichikawa
- Third Department of Internal Medicine, University of Fukui Faculty of Medical Sciences, 23-3, Matsuokashimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Tadashi Konoshita
- Third Department of Internal Medicine, University of Fukui Faculty of Medical Sciences, 23-3, Matsuokashimoaizuki, Eiheiji, Fukui, 910-1193, Japan.
| | - Yasukazu Makino
- Third Department of Internal Medicine, University of Fukui Faculty of Medical Sciences, 23-3, Matsuokashimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Jinya Suzuki
- Third Department of Internal Medicine, University of Fukui Faculty of Medical Sciences, 23-3, Matsuokashimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Tamotsu Ishizuka
- Third Department of Internal Medicine, University of Fukui Faculty of Medical Sciences, 23-3, Matsuokashimoaizuki, Eiheiji, Fukui, 910-1193, Japan
| | - Hiroyuki Nakamura
- Department of Environmental and Preventive Medicine, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| |
Collapse
|
34
|
Ethnic Prevalence of Angiotensin-Converting Enzyme Deletion (D) Polymorphism and COVID-19 Risk: Rationale for Use of Angiotensin-Converting Enzyme Inhibitors/Angiotensin Receptor Blockers. J Racial Ethn Health Disparities 2020; 8:973-980. [PMID: 32901433 PMCID: PMC7478439 DOI: 10.1007/s40615-020-00853-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/05/2020] [Accepted: 08/24/2020] [Indexed: 12/19/2022]
Abstract
Rationale Hypertension, obesity and diabetes are major risk factors associated with morbidities underlying COVID-19 infections. Regression analysis correlated presence of ACE insertion/deletion (I/D) polymorphism to COVID-19 incidence and mortality. Furthermore, COVID-19 prevalence correlated to allele frequency of angiotensin-converting enzyme (ACE) deletion (D) polymorphism within the European population. Objective Homozygous ACE deletion polymorphism is associated with increase in ACE and angiotensin II (Ang-II), sustained levels can result in inflammation, fibrosis and organ damage. The ACE DD polymorphism is also associated with hypertension, acute respiratory distress and diabetic nephropathy, all considered high risk for COVID-19 infection and outcomes. The study objective was to describe a biological framework associating ethnic prevalence of ACE deletion polymorphism to COVID-19 comorbidities providing rationale for therapeutic utility of ACE-I/ARBs to improve outcomes. Method and Results The Allele Frequency Database (ALFRED) was queried for frequency of rs4646994 representing ACE I/D polymorphism. In a total of 349 worldwide population samples, frequency of ACE D allele was higher in European, Asian, and Africans cohorts. In the USA, the frequency of ACE D allele was higher in non-Hispanic Black compared with non-Hispanic White and Mexican Americans. Conclusion COVID-19 binding mediated reduction/inactivation of ACE-II can increase ACE/Ang-II signalling pathway and related pathologies. The presence of ACE DD polymorphism with COVID-19 infection likely augments ACE/Ang-II activities, increasing severity of COVID-19 morbidities and impacts outcomes. Thus, ethnic prevalence of ACE DD polymorphism can explain in part the severity of COVID-19 morbidity providing rationale for the use of ACE-I/ARBs to improve outcomes.
Collapse
|
35
|
Tan HW, Xu Y, Lau ATY. Angiotensin-converting enzyme 2: The old door for new severe acute respiratory syndrome coronavirus 2 infection. Rev Med Virol 2020; 30:e2122. [PMID: 32602627 PMCID: PMC7361198 DOI: 10.1002/rmv.2122] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 04/30/2020] [Accepted: 05/04/2020] [Indexed: 02/05/2023]
Abstract
Coronavirus (CoV) disease 2019 (COVID-19) is an ongoing pandemic caused by severe acute respiratory syndrome CoV 2 (SARS-CoV-2). The highly contagious SARS-CoV-2 belongs to the genus Betacoronavirus, and it is phylogenetically closely related to SARS-CoV, a human CoV that caused an outbreak back in 2002 to 2003. Both SARS-CoV-2 and SARS-CoV enter human cells via the interactions between viral crown-like spike protein and human angiotensin-converting enzyme 2 (ACE2) receptor. Here, we aim to review the involvement of ACE2 in human CoV infections by discussing the roles of ACE2 in CoV evolution, cross-species transmissibility, and COVID-19 susceptibility. We also provide our perspectives on COVID-19 treatment and prevention.
Collapse
Affiliation(s)
- Heng Wee Tan
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and GeneticsShantou University Medical CollegeShantouGuangdongPeople's Republic of China
| | - Yan‐Ming Xu
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and GeneticsShantou University Medical CollegeShantouGuangdongPeople's Republic of China
| | - Andy T. Y. Lau
- Laboratory of Cancer Biology and Epigenetics, Department of Cell Biology and GeneticsShantou University Medical CollegeShantouGuangdongPeople's Republic of China
| |
Collapse
|
36
|
Zamai L. The Yin and Yang of ACE/ACE2 Pathways: The Rationale for the Use of Renin-Angiotensin System Inhibitors in COVID-19 Patients. Cells 2020; 9:E1704. [PMID: 32708755 PMCID: PMC7408073 DOI: 10.3390/cells9071704] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/10/2020] [Accepted: 06/25/2020] [Indexed: 02/06/2023] Open
Abstract
The article describes the rationale for inhibition of the renin-angiotensin system (RAS) pathways as specific targets in patients infected by SARS-CoV-2 in order to prevent positive feedback-loop mechanisms. Based purely on experimental studies in which RAS pathway inhibitors were administered in vivo to humans/rodents, a reasonable hypothesis of using inhibitors that block both ACE and ACE2 zinc metalloproteases and their downstream pathways in COVID-19 patients will be proposed. In particular, metal (zinc) chelators and renin inhibitors may work alone or in combination to inhibit the positive feedback loops (initially triggered by SARS-CoV-2 and subsequently sustained by hypoxia independently on viral trigger) as both arms of renin-angiotensin system are upregulated, leading to critical, advanced and untreatable stages of the disease.
Collapse
Affiliation(s)
- Loris Zamai
- Department of Biomolecular Sciences, University of Urbino “Carlo Bo”, 61032 Urbino, Italy; ; Tel.: +39-0722-304319
- INFN-Gran Sasso National Laboratory, Assergi, 67100 L’Aquila, Italy
| |
Collapse
|
37
|
Ghafouri-Fard S, Noroozi R, Omrani MD, Branicki W, Pośpiech E, Sayad A, Pyrc K, Łabaj PP, Vafaee R, Taheri M, Sanak M. Angiotensin converting enzyme: A review on expression profile and its association with human disorders with special focus on SARS-CoV-2 infection. Vascul Pharmacol 2020; 130:106680. [PMID: 32423553 PMCID: PMC7211701 DOI: 10.1016/j.vph.2020.106680] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/24/2020] [Accepted: 05/03/2020] [Indexed: 12/21/2022]
Abstract
Angiotensin-converting enzyme (ACE) and its homologue, ACE2, have been mostly associated with hypertensive disorder. However, recent pandemia of SARS-CoV-2 has put these proteins at the center of attention, as this virus has been shown to exploit ACE2 protein to enter cells. Clear difference in the response of affected patients to this virus has urged researchers to find the molecular basis and pathophysiology of the cell response to this virus. Different levels of expression and function of ACE proteins, underlying disorders, consumption of certain medications and the existence of certain genomic variants within ACE genes are possible explanations for the observed difference in the response of individuals to the SARS-CoV-2 infection. In the current review, we discuss the putative mechanisms for this observation.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Rezvan Noroozi
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Mir Davood Omrani
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Wojciech Branicki
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Ewelina Pośpiech
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Arezou Sayad
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Krzysztof Pyrc
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Paweł P Łabaj
- Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | - Reza Vafaee
- Proteomics Research Center, Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Taheri
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Marek Sanak
- Department of Internal Medicine, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
38
|
Abstract
PURPOSE OF THE REVIEW Angiotensin-converting enzyme 2 (ACE2) is a key counter-regulatory component of the renin-angiotensin system. Here, we briefly review the mechanistic and target organ effects related to ACE2 activity, and the importance of ACE2 in SARS-CoV-2 infection. RECENT FINDINGS ACE2 converts angiotensin (Ang) II to Ang-(1-7), which directly opposes the vasoconstrictive, proinflammatory, and prothrombotic effects of Ang II. ACE2 also facilitates SARS-CoV-2 viral entry into host cells. Drugs that interact with the renin-angiotensin system may impact ACE2 expression and COVID-19 pathogenesis; however, the magnitude and direction of these effects are unknown at this time. High quality research is needed to improve our understanding of how agents that act on the renin-angiotensin system impact ACE2 and COVID-19-related disease outcomes.
Collapse
Affiliation(s)
- Jordana B Cohen
- Renal-Electrolyte and Hypertension Division, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Thomas C Hanff
- Department of Biostatistics, Epidemiology, and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Adam P Bress
- Division of Health System Innovation and Research, Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
| | - Andrew M South
- Section of Nephrology, Department of Pediatrics, Wake Forest School of Medicine and Brenner Children's Hospital, Winston Salem, USA
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, Winston Salem, USA
- Department of Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine, Winston Salem, NC, USA
| |
Collapse
|
39
|
Pagliaro P, Penna C. ACE/ACE2 Ratio: A Key Also in 2019 Coronavirus Disease (Covid-19)? Front Med (Lausanne) 2020; 7:335. [PMID: 32626721 PMCID: PMC7314898 DOI: 10.3389/fmed.2020.00335] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 06/05/2020] [Indexed: 01/04/2023] Open
Affiliation(s)
- Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Torino, Turin, Italy
| | | |
Collapse
|
40
|
Samavati L, Uhal BD. ACE2, Much More Than Just a Receptor for SARS-COV-2. Front Cell Infect Microbiol 2020; 10:317. [PMID: 32582574 PMCID: PMC7294848 DOI: 10.3389/fcimb.2020.00317] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 05/26/2020] [Indexed: 12/17/2022] Open
Abstract
The rapidly evolving pandemic of severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection worldwide cost many lives. The angiotensin converting enzyme-2 (ACE-2) has been identified as the receptor for the SARS-CoV-2 viral entry. As such, it is now receiving renewed attention as a potential target for anti-viral therapeutics. We review the physiological functions of ACE2 in the cardiovascular system and the lungs, and how the activation of ACE2/MAS/G protein coupled receptor contributes in reducing acute injury and inhibiting fibrogenesis of the lungs and protecting the cardiovascular system. In this perspective, we predominantly focus on the impact of SARS-CoV-2 infection on ACE2 and dysregulation of the protective effect of ACE2/MAS/G protein pathway vs. the deleterious effect of Renin/Angiotensin/Aldosterone. We discuss the potential effect of invasion of SARS-CoV-2 on the function of ACE2 and the loss of the protective effect of the ACE2/MAS pathway in alveolar epithelial cells and how this may amplify systemic deleterious effect of renin-angiotensin aldosterone system (RAS) in the host. Furthermore, we speculate the potential of exploiting the modulation of ACE2/MAS pathway as a natural protection of lung injury by modulation of ACE2/MAS axis or by developing targeted drugs to inhibit proteases required for viral entry.
Collapse
Affiliation(s)
- Lobelia Samavati
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine and Detroit Medical Center, Wayne State University, Detroit, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bruce D. Uhal
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
41
|
Abstract
COVID-19 is the current public health threat all over the world. Unfortunately, there is no specific prevention and treatment strategy for this disease. We aim to explore the potential role of angiotensin-converting enzyme 2 (ACE2) in this regard through this literature review. As a crucial enzyme of renin-angiotensin-aldosterone system (RAAS), ACE2 not only mediates the virus entry but also affects the pathophysiological process of virus-induced acute lung injury (ALI), as well as other organs’ damage. As interaction of COVID-19 virus spike and ACE2 is essential for virus infection, COVID-19-specific vaccine based on spike protein, small molecule compound interrupting their interaction, human monoclonal antibody based on receptor-binding domain, and recombinant human ACE2 protein (rhuACE2) have aroused the interests of researchers. Meanwhile, ACE2 could catalyze angiotensin II (Ang II) to form angiotensin 1-7 (Ang 1-7), thus alleviates the harmful effect of Ang II and amplifies the protection effect of Ang1-7. ACE inhibitor and angiotensin II receptor blocker (ARB) have been shown to increase the level of expression of ACE2 and could be potential strategies in protecting lungs, heart, and kidneys. ACE2 plays a very important role in the pathogenesis and pathophysiology of COVID-19 infection. Strategies targeting ACE2 and its ligand, COVID-19 virus spike protein, may provide novel method in the prevention and management of novel coronavirus pneumonia.
Collapse
|
42
|
Samavati L, Uhal BD. ACE2, Much More Than Just a Receptor for SARS-COV-2. Front Cell Infect Microbiol 2020. [PMID: 32582574 DOI: 10.3389/fcimb.2020.0317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023] Open
Abstract
The rapidly evolving pandemic of severe acute respiratory syndrome coronavirus (SARS-CoV-2) infection worldwide cost many lives. The angiotensin converting enzyme-2 (ACE-2) has been identified as the receptor for the SARS-CoV-2 viral entry. As such, it is now receiving renewed attention as a potential target for anti-viral therapeutics. We review the physiological functions of ACE2 in the cardiovascular system and the lungs, and how the activation of ACE2/MAS/G protein coupled receptor contributes in reducing acute injury and inhibiting fibrogenesis of the lungs and protecting the cardiovascular system. In this perspective, we predominantly focus on the impact of SARS-CoV-2 infection on ACE2 and dysregulation of the protective effect of ACE2/MAS/G protein pathway vs. the deleterious effect of Renin/Angiotensin/Aldosterone. We discuss the potential effect of invasion of SARS-CoV-2 on the function of ACE2 and the loss of the protective effect of the ACE2/MAS pathway in alveolar epithelial cells and how this may amplify systemic deleterious effect of renin-angiotensin aldosterone system (RAS) in the host. Furthermore, we speculate the potential of exploiting the modulation of ACE2/MAS pathway as a natural protection of lung injury by modulation of ACE2/MAS axis or by developing targeted drugs to inhibit proteases required for viral entry.
Collapse
Affiliation(s)
- Lobelia Samavati
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, School of Medicine and Detroit Medical Center, Wayne State University, Detroit, MI, United States
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, United States
| | - Bruce D Uhal
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
43
|
Zhang YY, Yu Y, Yu C. Antifibrotic Roles of RAAS Blockers: Update. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1165:671-691. [PMID: 31399990 PMCID: PMC7121580 DOI: 10.1007/978-981-13-8871-2_33] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The rennin-angiotensin-aldosterone system (RAAS) has been well documented in regulating blood pressure, fluid volume, and sodium balance. Overactivity of RAAS promotes both systemic and regional glomerular capillary hypertension, which could induce hemodynamic injury to the glomerulus, leading to kidney damage and renal fibrosis via profibrotic and proinflammatory pathway. Therefore, the use of RAAS inhibitors (i.e., ACEIs, ARBs, and MRAs) as the optional therapy has been demonstrated to prevent proteinuria, and kidney fibrosis and slow the decline of renal function effectively in the process of kidney disease during the last few decades. Recently, several new components of the RAAS have been discovered, including ACE2 and the corresponding ACE2/Ang (1-7)/Mas axis, which are also present in the kidney. Besides the classic RAAS inhibitors target the angiotensin-AT1-aldosterone axis, with the expanding knowledge about RAAS, a number of potential therapeutic targets in this system is emerging. Newer agents that are more specific are being developed. The present chapter outlines the insights of the RAAS agents (classic RAAS antagonists/the new RAAS drugs), and discusses its clinical application in the combat of renal fibrosis.
Collapse
Affiliation(s)
- Ying-Ying Zhang
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ying Yu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chen Yu
- Department of Nephrology, Tongji Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
44
|
Wang Z, Wang S, Zhao J, Yu C, Hu Y, Tu Y, Yang Z, Zheng J, Wang Y, Gao Y. Naringenin Ameliorates Renovascular Hypertensive Renal Damage by Normalizing the Balance of Renin-Angiotensin System Components in Rats. Int J Med Sci 2019; 16:644-653. [PMID: 31217731 PMCID: PMC6566737 DOI: 10.7150/ijms.31075] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/07/2019] [Indexed: 01/08/2023] Open
Abstract
Background: Naringenin, a member of the dihydroflavone family, has been shown to have a protective function in multiple diseases. We previously demonstrated that naringenin played a protective role in hypertensive myocardial hypertrophy by decreasing angiotensin-converting enzyme (ACE) expression. The kidney is a primary target organ of hypertension. The present study tested the effect of naringenin on renovascular hypertensive kidney damage and explored the underlying mechanism. Methods and Results: An animal model of renovascular hypertension was established by performing 2-kidney, 1-clip (2K1C) surgery in Sprague Dawley rats. Naringenin (200 mg/kg/day) or vehicle was administered for 10 weeks. Blood pressure and urinary protein were continuously monitored. Plasma parameters, renal pathology and gene expression of nonclipped kidneys were evaluated by enzyme-linked immunosorbent assay, histology, immunohistochemistry, real-time polymerase chain reaction, and Western blot at the end of the study. Rats that underwent 2K1C surgery exhibited marked elevations of blood pressure and plasma Ang II levels and renal damage, including mesangial expansion, interstitial fibrosis, and arteriolar thickening in the nonclipped kidneys. Naringenin significantly ameliorated hypertensive nephropathy and retarded the rise of Ang II levels in peripheral blood but had no effect on blood pressure. 2K1C rats exhibited increases in the ACE/ACE2 protein ratio and AT1R/AT2R protein ratio in the nonclipped kidney compared with sham rats, and these increases were significantly suppressed by naringenin treatment. Conclusions: Naringenin attenuated renal damage in a rat model of renovascular hypertension by normalizing the imbalance of renin-angiotensin system activation. Our results suggest a potential treatment strategy for hypertensive nephropathy.
Collapse
Affiliation(s)
- Zhizhi Wang
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, China
| | - Shanshan Wang
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, China
| | - Jianqiao Zhao
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Changan Yu
- Central Laboratory of Cardiovascular Disease, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yi Hu
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, China
| | - Yimin Tu
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Zufang Yang
- Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China
| | - Jingang Zheng
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, China.,Department of Cardiology, Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China.,Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yong Wang
- Department of Cardiology, China-Japan Friendship School of Clinical Medicine, Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, 100029, China.,Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yanxiang Gao
- Department of Cardiology, China-Japan Friendship Hospital, Beijing 100029, China
| |
Collapse
|
45
|
Yang CW, Lu LC, Chang CC, Cho CC, Hsieh WY, Tsai CH, Lin YC, Lin CS. Imbalanced plasma ACE and ACE2 level in the uremic patients with cardiovascular diseases and its change during a single hemodialysis session. Ren Fail 2018; 39:719-728. [PMID: 29157100 PMCID: PMC6446170 DOI: 10.1080/0886022x.2017.1398665] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background: The renin-angiotensin system (RAS) has significant influences on heart and renal disease progression. Angiotensin converting enzyme (ACE) and angiotensin converting enzyme II (ACE2) are major peptidases of RAS components and play counteracting functions through angiotensin II (Ang II)/ATIR and angiotensin-(1–7) (Ang-(1–7))/Mas axis, respectively. Methods: There were 360 uremic patients on regular hemodialysis (HD) treatment (inclusive of 119 HD patients with cardiovascular diseases (CVD) and 241 HD patients without CVD and 50 healthy subjects were enrolled in this study. Plasma ACE, ACE2, Ang II and Ang-(1–7) levels of the HD patients were determined. Results: We compared pre-HD levels of plasma ACE, ACE2, Ang II and Ang-(1–7) in the HD patients with and without CVD to those of the controls. The HD patients, particularly those with CVD, showed a significant increase in the levels of ACE and Ang II, whereas ACE2 and Ang-(1–7) levels were lower than those in the healthy controls. Therefore, imbalanced ACE/ACE2 was observed in the HD patients with CVD. In the course of a single HD session, the plasma ACE, ACE/ACE2 and Ang II levels in the HD patients with CVD were increased from pre-HD to post-HD. On the contrary, ACE2 levels were decreased after the HD session. These changes were not detected in the HD patients without CVD. Conclusions: Pathogenically imbalanced circulating ACE/ACE2 was detected in the HD patients, particularly those with CVD. HD session could increase ACE/Ang II/AT1R axis and decrease ACE2/Ang-(1–7)/Mas axis activity in the circulation of HD patients with CVD.
Collapse
Affiliation(s)
- Chung-Wei Yang
- a Department of Biological Science and Technology , National Chiao Tung University , Hsinchu , Taiwan.,b Division of Nephrology, Department of Internal Medicine , National Taiwan University Hospital Hsinchu Branch , Hsinchu , Taiwan
| | - Li-Che Lu
- c Division of Nephrology, Department of Internal Medicine , Shin Kong Wu Ho-Su Memorial Hospital , Taipei , Taiwan
| | - Chia-Chu Chang
- d Division of Nephrology, Department of Internal Medicine , Changhua Christian Hospital , Changhua , Taiwan.,e School of Medicine , Chung-Shan Medical University , Taichung , Taiwan
| | - Ching-Chang Cho
- a Department of Biological Science and Technology , National Chiao Tung University , Hsinchu , Taiwan
| | - Wen-Yeh Hsieh
- f Division of Chest Medicine, Department of Internal Medicine , Hsinchu Mackay Memorial Hospital , Hsinchu , Taiwan.,g Department of Senior Citizen Service Management , Minghsin University of Science and Technology , Hsinchu , Taiwan
| | - Chin-Hung Tsai
- a Department of Biological Science and Technology , National Chiao Tung University , Hsinchu , Taiwan
| | - Yi-Chang Lin
- a Department of Biological Science and Technology , National Chiao Tung University , Hsinchu , Taiwan
| | - Chih-Sheng Lin
- a Department of Biological Science and Technology , National Chiao Tung University , Hsinchu , Taiwan
| |
Collapse
|
46
|
Spironolactone rescues renal dysfunction in obstructive jaundice rats by upregulating ACE2 expression. J Cell Commun Signal 2018; 13:17-26. [PMID: 29882088 DOI: 10.1007/s12079-018-0466-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 04/17/2018] [Indexed: 12/31/2022] Open
Abstract
Postoperative acute renal failure in patients with obstructive jaundice is still a serious clinically complication, yet the mechanisms remain unclear. Renin-angiotensin-aldosterone system (RAAS) plays a central role in renal disease progression. Several lines of evidence shows that angiotensin-converting-enzyme-2 (ACE2), a main effector of RAAS acts as a local regulator for renal protection. This study aims to investigate the role of ACE2 and the effect of spironolactone treatment in obstructive jaundice(OJ) rats with renal injury. The rats with obstructive jaundice were established by bile duct ligation. Total bilirubin (TBil), serum creatinine (Scr) and the expression of ACE2 in kidney tissue of obstructive jaundice rats were detected. Comparatively, the expression of ACE2, renin, angiotensin II (AngII), angiotensin-(1-7)[Ang-(1-7)], aldosterone and intercellular adhesion molecule 1 (ICAM-1) in kidney tissues after spironolactone administration were measured by ELISA. Renal necrosis, inflammation and fibrosis induced by OJ were also measured by HE staining and Masson staining. The correlation between the expression of ACE2 and TBil, also the Scr level were investigated. With the time of common bile duct ligation prolonged, the TBil and Scr concentration increased while the expression of ACE2 in OJ rats' kidney tissues decreased. However, after spironolactone intervention, the expressions of ACE2, renin, AngII, Ang-(1-7), aldosterone and ICAM-1 in kidney tissue were changed, moreover, necrotic, inflammatory and fibrotic condition was also decreased. The relationship between the mRNA expression of ACE2 and TBil/Scr was observed to be moderately negatively correlated (r = -0.516, R2 = 0.292, P < 0.01), (r = -0.576, R2 = 0.332, P < 0.01), respectively. RAAS exerted an important effect in the renal damage caused by OJ. Spironolactone intervention not only improved the degree of renal fibrosis induced by OJ, but also upregulated the ACE2 expression in the kidney of OJ rats and rescued the renal function.
Collapse
|
47
|
Xu X, Cai Y, Yu Y. Effects of a novel curcumin derivative on the functions of kidney in streptozotocin-induced type 2 diabetic rats. Inflammopharmacology 2018; 26:1257-1264. [PMID: 29582239 PMCID: PMC6153927 DOI: 10.1007/s10787-018-0449-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 02/01/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE B6, an analog of curcumin, is a compound isolated from a traditional Chinese medicine Turmeric. In this paper, we aimed to explore the efficacy of B6 on diabetic nephropathy and the related mechanisms. MATERIALS AND METHODS The effects of B6 were studied on fast-blood glucose, serum creatinine, urea nitrogen, urine albumen/24 h, pathological changes of main organs, the levels of ACE2 and ACE2 mRNA in the rat model of diabetes induced by streptozotocin. RESULTS The results showed that B6 treatment could reduce serum creatinine, urea nitrogen, urine albumen/24 h, decrease the level of AngII, improve the renal pathological changes in diabetic rats and increase the levels of ACE2 and ACE2 mRNA. CONCLUSION These results suggested B6 could protect the renal function of diabetic rats. This study provided scientific basis for the further researches and clinical applications of B6.
Collapse
Affiliation(s)
- Xuegu Xu
- Department of Pharmacy, The Eye Hospital of Wenzhou Medical University, Wenzhou, 325003, Zhejiang, China
| | - Yonghao Cai
- Department of Pharmacy, The Eye Hospital of Wenzhou Medical University, Wenzhou, 325003, Zhejiang, China
| | - Yinfei Yu
- Department of Pharmacy, The Eye Hospital of Wenzhou Medical University, Wenzhou, 325003, Zhejiang, China.
| |
Collapse
|
48
|
Gaur P, Saini S, Vats P, Kumar B. Regulation, signalling and functions of hormonal peptides in pulmonary vascular remodelling during hypoxia. Endocrine 2018; 59:466-480. [PMID: 29383676 DOI: 10.1007/s12020-018-1529-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 01/10/2018] [Indexed: 01/06/2023]
Abstract
Hypoxic state affects organism primarily by decreasing the amount of oxygen reaching the cells and tissues. To adjust with changing environment organism undergoes mechanisms which are necessary for acclimatization to hypoxic stress. Pulmonary vascular remodelling is one such mechanism controlled by hormonal peptides present in blood circulation for acclimatization. Activation of peptides regulates constriction and relaxation of blood vessels of pulmonary and systemic circulation. Thus, understanding of vascular tone maintenance and hypoxic pulmonary vasoconstriction like pathophysiological condition during hypoxia is of prime importance. Endothelin-1 (ET-1), atrial natriuretic peptide (ANP), and renin angiotensin system (RAS) function, their receptor functioning and signalling during hypoxia in different body parts point them as disease markers. In vivo and in vitro studies have helped understanding the mechanism of hormonal peptides for better acclimatization to hypoxic stress and interventions for better management of vascular remodelling in different models like cell, rat, and human is discussed in this review.
Collapse
Affiliation(s)
- Priya Gaur
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| | - Supriya Saini
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| | - Praveen Vats
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India.
| | - Bhuvnesh Kumar
- Defence Institute of Physiology and Allied Sciences, Lucknow Road, Timarpur, Delhi, India
| |
Collapse
|
49
|
On the top of ARB N/L type Ca channel blocker leads to less elevation of aldosterone. Biosci Rep 2016; 36:BSR20160129. [PMID: 27515419 PMCID: PMC5025805 DOI: 10.1042/bsr20160129] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/11/2016] [Indexed: 01/04/2023] Open
Abstract
The activation of the renin–angiotensin system (RAS) is one of the unfavourable characteristics of calcium channel blocker (CCB). N type calcium channel is thought to be involved in renin gene transcription and adrenal aldosterone release. Accordingly, N/L type CCB has a possibility of less elevation of plasma aldosterone concentrations (PAC) among CCBs. In a monotherapy study, we had already demonstrated that N/L type CCB leads to less activation of the RAS compared with L type CCB. The objective of this study is to substantiate the hypothesis that at the condition of additive administration on the top of an angiotensin receptor blocker (ARB), still N/L type CCB leads to less elevation of PAC compared with L type one. Subjects were 60 hypertensives administered with valsartan. As an open label study, amlodipine (L type) or cilnidipine (N/L type) were administered on the top of valsartan (ARB) in a cross-over manner. Results were as follows (valsartan+amlodipine compared with valsartan+cilnidipine): systolic blood pressure (SBP)/diastolic blood pressure (DBP) (mmHg): 132±10/76±10 compared with 131±10/77±9, P=0.95/0.48, plasma renin activity (PRA) (ng/ml·h): 2.41±2.67 compared with 2.00±1.50 P=0.20, PAC (pg/ml): 77.3±31.0 compared with 67.4±24.8, P<0.05, urinary albumin excretion (UAE) (mg/gCr): 105.9±216.1 compared with 73.9±122.2, P<0.05. Thus, PAC at cilnidipine was significantly lower than those at amlodipine in spite of the comparable BP reductions. Besides, UAE was significantly lower at cilnidipine. In conclusion, on the top of the ARB, it is suggested that cilnidipine administration might lead to less elevation of PAC and reduction in UAE compared with amlodipine.
Collapse
|
50
|
Gowrisankar YV, Clark MA. Angiotensin II regulation of angiotensin-converting enzymes in spontaneously hypertensive rat primary astrocyte cultures. J Neurochem 2016; 138:74-85. [PMID: 27085714 DOI: 10.1111/jnc.13641] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Revised: 03/03/2016] [Accepted: 03/23/2016] [Indexed: 02/07/2023]
Abstract
Angiotensin (Ang) II plays a critical role in cardiovascular and blood pressure regulation. Ang II is produced by angiotensin-converting enzyme (ACE) and it interacts with the Ang AT1 receptor to cause much of its well-known cardiovascular effects. Ang-(1-7) is another active peptide produced by the rennin-angiotensin system. This peptide is produced from Ang I or Ang II by the catalytic activity of ACE2. Ang-(1-7) interacts with the Mas receptor to counteract many of the effects of Ang II. Thus, the ACE2/Ang-(1-7)/Mas axis acts opposite of the ACE/Ang II/AT1 axis. In this study we investigated how Ang II regulates the key enzymes of these axes, ACE and its homolog ACE2, and determined whether they are dysregulated in the hypertensive condition. Brainstem and cerebellum astrocytes isolated from the spontaneously hypertensive rat (SHR) were used in these studies. Ang II effect on the enzymes' mRNA and protein levels was measured using quantitative PCR and western blotting techniques, respectively. Results from this study showed that Ang II up-regulated ACE protein levels, but down-regulated ACE mRNA levels in brainstem and cerebellum astrocytes in both models. Ang II also reduced ACE2 mRNA expression in SHR and Wistar astrocytes isolated from both brain regions. Ang II effects on ACE2 protein were biphasic. In SHR astrocytes, Ang II-mediated ACE2 protein initially increased then decreased at later time points. In contrast, in Wistar astrocytes, Ang II initially decreased ACE2 protein expression, but up-regulated the protein at later time points. The findings of these studies suggest that Ang II has a differential effect on ACE and ACE2 expression. Furthermore, in the SHR model there may be alteration in the ACE/ACE2 balance in a manner that favors increased Ang II generation and decreased Ang-(1-7) production contributing to the hypertensive phenotype observed in this model. The levels of angiotensin (Ang) II depend on the actions of angiotensin-converting enzyme (ACE) and ACE2. We showed in astrocytes isolated from the SHRs that Ang II differentially affects ACE and ACE2 expression. There may be an alteration in the ACE/ACE2 balance favoring Ang II generation. This imbalance may contribute to the hypertensive phenotype observed in this SHR model.
Collapse
Affiliation(s)
- Yugandhar V Gowrisankar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| | - Michelle A Clark
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, Florida, USA
| |
Collapse
|