1
|
Lissoni P, Rovelli F, Messina G, Monzon A, Valentini A, Sassola A, Di Fede G, Simoes-de-Silva AC, Merli N, Bartsch C, Vlaescu VG, Cardinali DD. Psycho-Neuro-EndocrinE-Immunology Therapy of Cancer, Autoimmunity, Geriatric Disorders, Covid-19, and Hypertension. Methods Mol Biol 2025; 2868:111-132. [PMID: 39546228 DOI: 10.1007/978-1-0716-4200-9_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
Despite the great number of experimental investigations in the area of psycho-neuro-endocrine-immunology showing that endocrine, nervous, and immune systems cannot be in vivo physiologically separated, the diagnosis and therapies of the pathologies of these three functional biological systems continue to be separately performed from a clinical practice point of view. The separation between experimental and clinical medicine became dramatic after the discovery of more than 10 human molecules provided by anti-inflammatory and antitumor activity, completely devoid of any toxicity, which may be subdivided into three fundamental classes, consisting of the pineal indole, beta-carboline, and methoxy-kynuramine hormones. Moreover, human systemic diseases, including cancer, autoimmunity, and cardiovascular pathologies, despite their different pathogenesis and symptomatology, are commonly characterized by a progressive decline in the endogenous production of pineal hormones, endocannabinoids, and Ang 1-7, with a consequent inflammatory status and diminished natural resistance against cancer. Then the evaluation of the functional status of the pineal gland, the endocannabinoid system, and ACE2-Ang 1-7 axis should have to be included within the laboratory analyses for the systemic diseases. Finally, the correction of cancer- and autoimmunity-related neuroimmune and neuroendocrine alterations could influence the clinical course of systemic diseases. In fact, preliminary clinical results would demonstrate that the neuroimmune regimen with pineal hormones, cannabinoids, and Ang 1-7 may allow clinical benefits also in patients affected by systemic pathologies, including cancer, autoimmunity, and cardiovascular diseases, who did not respond to the standard therapies.
Collapse
Affiliation(s)
- Paolo Lissoni
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Franco Rovelli
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Giusy Messina
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Alejandra Monzon
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | - Agnese Valentini
- Institute of Biological Medicine, Milan, Italy
- Madonna del Soccorso Hospital, San Benedetto del Tronto, Ascoli Piceno, Italy
| | | | | | | | | | | | | | | |
Collapse
|
2
|
Takeshita H, Yamamoto K, Mogi M, Rakugi H. Muscle mass, muscle strength and the renin-angiotensin system. Clin Sci (Lond) 2024; 138:1561-1577. [PMID: 39718491 DOI: 10.1042/cs20220501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 11/19/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024]
Abstract
The renin-angiotensin system (RAS) is a classically known circulatory regulatory system. In addition to the previously known multi-organ circulatory form of the RAS, the existence of tissue RASs in individual organs has been well established. Skeletal muscle has also been identified as an organ with a distinct RAS. In recent years, the effects of RAS activation on skeletal muscle have been elucidated from several perspectives: differences in motor function due to genetic polymorphisms of RAS components, skeletal muscle dysfunction under conditions of excessive RAS activation such as heart failure, and the effects of the use of RAS inhibitors on muscle strength. In addition, the concept of the RAS itself has recently been expanded with the discovery of a 'protective arm' of the RAS formed by factors such as angiotensin-converting enzyme 2 and angiotensin 1-7. This has led to a new understanding of the physiological function of the RAS in skeletal muscle. This review summarizes the diverse physiological functions of the RAS in skeletal muscle and considers the potential of future therapeutic strategies targeting the RAS to overcome problems such as sarcopenia and muscle weakness associated with chronic disease.
Collapse
Affiliation(s)
- Hikari Takeshita
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Department of Medical Science Technology, Faculty of Medical Science Technology, Morinomiya University of Medical Sciences, Osaka, Osaka, Japan
| | - Koichi Yamamoto
- Department of Geriatric and General Medicine, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masaki Mogi
- Department of Pharmacology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | | |
Collapse
|
3
|
Duan R, Shi L, Deng Y, Wu J, Wang S, Peng Q, Li Z, Xu Z, Wang F, Xue X, Gao Q. AVE0991 ameliorates dopaminergic neuronal damage in Parkinson's disease through HOTAIRM1/miR-223-3p/α-synuclein axis. Sci Rep 2024; 14:26346. [PMID: 39487232 PMCID: PMC11530439 DOI: 10.1038/s41598-024-76058-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/10/2024] [Indexed: 11/04/2024] Open
Abstract
Parkinson's disease (PD) is a prevalent type of neurodegenerative disorder. AVE0991, a non-peptide analogue of Ang-(1-7), by which the progression of PD has been discovered to be ameliorated, but the specific mechanism whereby AVE0991 modulates the progression of PD re-mains unclear. The mice overexpressing human α-syn (A53T) were established to simulate PD pathology, and we also constructed an in vitro model of mouse dopaminergic neurons overexpressing hα-syn (A53T). The [18F] FDG-PET/CT method was employed to assess FDG uptake in human α-syn (A53T) overexpressing mice. Levels of lnc HOTAIRM1 and miR-223-3p were detected via qRT-PCR. Flow cytometry was deployed to assay cell apoptosis. Here, we found that AVE0991 improved behaviour disorders and decreased α-syn expression in the substantia nigra of mice with Parkinson's disease. AVE0991 inhibited the apoptosis of dopaminergic neurons overexpressing hα-syn (A53T) via lncRNA HOTAIRM1. MiR-223-3p binds to HOTAIRM1 as a ceRNA and directly targets α-syn. Moreover, miR-223-3p level in peripheral blood was found negatively correlated with the α-syn. Our present study shows that the angiotensin-(1-7) analogue AVE0991 targeted at the HOTAIRM1/miR-223-3p axis to degrade α-synuclein in PD mice, and showed neuroprotection in vitro.
Collapse
Affiliation(s)
- Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Liang Shi
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Yang Deng
- School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Jiang Wu
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Shiyao Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Qiang Peng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Zhongyuan Li
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Zhaohan Xu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China
| | - Feng Wang
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Xue Xue
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| | - Qing Gao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
- Department of Nuclear Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, China.
| |
Collapse
|
4
|
Nakano T, Goto S, Chen CL. Mechanisms of Tolerance Induction in Liver Transplantation: Lessons Learned from Fetomaternal Tolerance, Autoimmunity and Tumor Immunity. Int J Mol Sci 2024; 25:9331. [PMID: 39273280 PMCID: PMC11395488 DOI: 10.3390/ijms25179331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/08/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Since the first published report of experimental kidney transplantation in dogs in 1902, there were many experimental and clinical trials of organ transplantation, with many sacrifices. After the establishment of the surgical technique and the discovery of immunosuppressive drugs, transplantation became the definitive treatment strategy for patients with terminal organ failure. However, this is not a common therapy method due to the difficulty of solving the fundamental issues behind organ transplantation, including the shortage of donor graft, potential risks of transplant surgery and economic capability. The pre- and post-transplant management of recipients is another critical issue that may affect transplant outcome. Most liver transplant recipients experience post-transplant complications, including infection, acute/chronic rejection, metabolic syndrome and the recurrence of hepatocellular carcinoma. Therefore, the early prediction and diagnosis of these complications may improve overall and disease-free survival. Furthermore, how to induce operational tolerance is the key to achieving the ultimate goal of transplantation. In this review, we focus on liver transplantation, which is known to achieve operational tolerance in some circumstances, and the mechanical similarities and differences between liver transplant immunology and fetomaternal tolerance, autoimmunity or tumor immunity are discussed.
Collapse
Affiliation(s)
- Toshiaki Nakano
- Graduate Institute of Clinical Medical Sciences, Chang Gung University College of Medicine, Taoyuan 333, Taiwan
- Liver Transplantation Center and Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| | - Shigeru Goto
- Liver Transplantation Center and Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Nobeoka Medical Check Center, Fukuoka Institution of Occupational Health, Nobeoka 882-0872, Japan
- School of Pharmacy, Shujitsu University, Okayama 703-8516, Japan
| | - Chao-Long Chen
- Liver Transplantation Center and Department of Surgery, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
| |
Collapse
|
5
|
da Silva WN, Carvalho Costa PA, Scalzo Júnior SRA, Ferreira HAS, Prazeres PHDM, Campos CLV, Rodrigues Alves MT, Alves da Silva NJ, de Castro Santos AL, Guimarães LC, Chen Ferris ME, Thatte A, Hamilton A, Bicalho KA, Lobo AO, Santiago HDC, da Silva Barcelos L, Figueiredo MM, Teixeira MM, Vasconcelos Costa V, Mitchell MJ, Frézard F, Pires Goulart Guimaraes P. Ionizable Lipid Nanoparticle-Mediated TRAIL mRNA Delivery in the Tumor Microenvironment to Inhibit Colon Cancer Progression. Int J Nanomedicine 2024; 19:2655-2673. [PMID: 38500680 PMCID: PMC10946446 DOI: 10.2147/ijn.s452896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 03/05/2024] [Indexed: 03/20/2024] Open
Abstract
Introduction Immunotherapy has revolutionized cancer treatment by harnessing the immune system to enhance antitumor responses while minimizing off-target effects. Among the promising cancer-specific therapies, tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has attracted significant attention. Methods Here, we developed an ionizable lipid nanoparticle (LNP) platform to deliver TRAIL mRNA (LNP-TRAIL) directly to the tumor microenvironment (TME) to induce tumor cell death. Our LNP-TRAIL was formulated via microfluidic mixing and the induction of tumor cell death was assessed in vitro. Next, we investigated the ability of LNP-TRAIL to inhibit colon cancer progression in vivo in combination with a TME normalization approach using Losartan (Los) or angiotensin 1-7 (Ang(1-7)) to reduce vascular compression and deposition of extracellular matrix in mice. Results Our results demonstrated that LNP-TRAIL induced tumor cell death in vitro and effectively inhibited colon cancer progression in vivo, particularly when combined with TME normalization induced by treatment Los or Ang(1-7). In addition, potent tumor cell death as well as enhanced apoptosis and necrosis was found in the tumor tissue of a group treated with LNP-TRAIL combined with TME normalization. Discussion Together, our data demonstrate the potential of the LNP to deliver TRAIL mRNA to the TME and to induce tumor cell death, especially when combined with TME normalization. Therefore, these findings provide important insights for the development of novel therapeutic strategies for the immunotherapy of solid tumors.
Collapse
Affiliation(s)
- Walison Nunes da Silva
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Heloísa A S Ferreira
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | | | | | | | - Lays Cordeiro Guimarães
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Maria Eduarda Chen Ferris
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Ajay Thatte
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Alex Hamilton
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Helton da Costa Santiago
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Lucíola da Silva Barcelos
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Mauro Martins Teixeira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | | | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Frédéric Frézard
- Department of Physiology and Biophysics, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | |
Collapse
|
6
|
Zorad S, Skrabanova M, Zilkova M, Cente M, Turic Csokova N, Kovacech B, Cizkova D, Filipcik P. Angiotensin I and II Stimulate Cell Invasion of SARS-CoV-2: Potential Mechanism via Inhibition of ACE2 Arm of RAS. Physiol Res 2024; 73:27-35. [PMID: 38466002 PMCID: PMC11019619 DOI: 10.33549/physiolres.935198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/27/2023] [Indexed: 04/26/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2), one of the key enzymes of the renin-angiotensin system (RAS), plays an important role in SARS-CoV-2 infection by functioning as a virus receptor. Angiotensin peptides Ang I and Ang II, the substrates of ACE2, can modulate the binding of SARS-CoV-2 Spike protein to the ACE2 receptor. In the present work, we found that co incubation of HEK-ACE2 and Vero E6 cells with the SARS-CoV-2 Spike pseudovirus (PVP) resulted in stimulation of the virus entry at low and high micromolar concentrations of Ang I and Ang II, respectively. The potency of Ang I and Ang II stimulation of virus entry corresponds to their binding affinity to ACE2 catalytic pocket with 10 times higher efficiency of Ang II. The Ang II induced mild increase of PVP infectivity at 20 microM; while at 100 microM the increase (129.74+/-3.99 %) was highly significant (p<0.001). Since the angiotensin peptides act in HEK ACE2 cells without the involvement of angiotensin type I receptors, we hypothesize that there is a steric interaction between the catalytic pocket of the ACE2 enzyme and the SARS-CoV-2 S1 binding domain. Oversaturation of the ACE2 with their angiotensin substrate might result in increased binding and entry of the SARS-CoV-2. In addition, the analysis of angiotensin peptides metabolism showed decreased ACE2 and increased ACE activity upon SARS-CoV-2 action. These effects should be taken into consideration in COVID-19 patients suffering from comorbidities such as the over-activated renin-angiotensin system as a mechanism potentially influencing the SARS-CoV-2 invasion into recipient cells.
Collapse
Affiliation(s)
- S Zorad
- Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovak Republic. and Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | | | | | | | | | | | | | |
Collapse
|
7
|
Nakhaie S, Yazdani R, Shakibi M, Torabian S, Pezeshki S, Bazrafshani MS, Azimi M, Salajegheh F. The effects of antihypertensive medications on severity and outcomes of hypertensive patients with COVID-19. J Hum Hypertens 2023; 37:511-518. [PMID: 35790875 PMCID: PMC9255835 DOI: 10.1038/s41371-022-00716-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/19/2022] [Accepted: 06/14/2022] [Indexed: 02/03/2023]
Abstract
In Covid-19 pandemic, specific comorbidities are associated with the increased risk of worse outcomes and increased severity of lung injury and mortality. the aim of this study was to investigate the effects of antihypertensive medications on the severity and outcomes of hypertensive patients with COVID-19. This retrospective observational study conducted on patients with COVID-19 who referred to Afzalipour Hospital, Kerman, Iran during the six months from 19 February 2020 to 20 July 2020. The data were collected through medical chart reviews. We assessed 265 patients with Covid-19 and they stratified based on hypertension and type of antihypertension medications. The data were described and Student's t-test, Mann-Whitney U and Fisher exact test were run to compare the patients 'demographical and clinical information. The qualitative variables were compared using the by SPSS software version 23. The results of the present study showed that hypertension was a prevalent comorbidity among patients with COVID-19 and hypertensive patients compared to other patients without any comorbidity who were older (P-value: 0.03). The oxygen saturation was higher for the patients in the control group than hypertensive patients (P-value: 0.01). The severity of COVID-19 and its outcome were not different between the patients who took or did not take antihypertensive medications and also the type of antihypertensive medications. Hypertensive patients did not show any significant difference in survival, hospital stay, ICU admission, disease severity, and invasive medical ventilation in other normotensive patients with COVID-19.
Collapse
Affiliation(s)
- Samira Nakhaie
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Rostam Yazdani
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammadreza Shakibi
- HIV/STI Surveillance Research Center, and WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Soheila Torabian
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Sara Pezeshki
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran
| | - Maliheh Sadat Bazrafshani
- HIV/STI Surveillance Research Center, and WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Azimi
- Department of Traditional Medicine, School of Traditional Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Faranak Salajegheh
- Endocrinology and Metabolism Research Center, Institute of Basic and Clinical Physiology Sciences, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
8
|
Abstract
The current epidemic of corona virus disease (COVID-19) has resulted in an immense health burden that became the third leading cause of death and potentially contributed to a decline in life expectancy in the United States. The severe acute respiratory syndrome-related coronavirus-2 binds to the surface-bound peptidase angiotensin-converting enzyme 2 (ACE2, EC 3.4.17.23) leading to tissue infection and viral replication. ACE2 is an important enzymatic component of the renin-angiotensin system (RAS) expressed in the lung and other organs. The peptidase regulates the levels of the peptide hormones Ang II and Ang-(1-7), which have distinct and opposing actions to one another, as well as other cardiovascular peptides. A potential consequence of severe acute respiratory syndrome-related coronavirus-2 infection is reduced ACE2 activity by internalization of the viral-ACE2 complex and subsequent activation of the RAS (higher ratio of Ang II:Ang-[1-7]) that may exacerbate the acute inflammatory events in COVID-19 patients and possibly contribute to the effects of long COVID-19. Moreover, COVID-19 patients present with an array of autoantibodies to various components of the RAS including the peptide Ang II, the enzyme ACE2, and the AT1 AT2 and Mas receptors. Greater disease severity is also evident in male COVID-19 patients, which may reflect underlying sex differences in the regulation of the 2 distinct functional arms of the RAS. The current review provides a critical evaluation of the evidence for an activated RAS in COVID-19 subjects and whether this system contributes to the greater severity of severe acute respiratory syndrome-related coronavirus-2 infection in males as compared with females.
Collapse
Affiliation(s)
- Mark C. Chappell
- Hypertension and Vascular Research Center, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
9
|
Kim Y, Kim J, Han SJ. Diminazene aceturate exacerbates renal fibrosis after unilateral ureteral obstruction in female mice. Kidney Res Clin Pract 2023; 42:188-201. [PMID: 37037481 PMCID: PMC10085718 DOI: 10.23876/j.krcp.22.033] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 06/07/2022] [Indexed: 04/03/2023] Open
Abstract
Background: Diminazene aceturate (DIZE), an angiotensin-converting enzyme 2 (ACE2) activator, exerts anti-inflammatory and antifibrotic effects in a variety of human chronic diseases. However, the role of DIZE in kidney fibrosis and the underlying mechanism remain unclear. Therefore, we investigated the effects of DIZE on the progression of renal fibrosis after unilateral ureteral obstruction (UUO), a well-established model of chronic kidney disease. Methods: C57BL/6 female or male mice were subjected to right UUO. Mice received 15 mg/kg DIZE or vehicle (saline) daily. On the 7th day after UUO, kidneys were collected for analysis of renal fibrosis (α-smooth muscle actin, phosphorylated SMAD3, transforming growth factor (TGF)-β, Masson’s trichrome, and Sirius red staining), inflammation (macrophage infiltration, proinflammatory cytokines/chemokines), apoptosis/necrotic cell death (TUNEL and periodic acid-Schiff staining), and ACE2 activity and messenger RNA (mRNA) expression.Results: Treatment with DIZE exacerbated renal fibrosis by upregulating the profibrotic TGF-β/SMAD3 pathway, proinflammatory cytokine/chemokines (interleukin [IL]-1β, monocyte chemoattractant protein-1, IL-6, and macrophage inflammatory protein-2) levels, M2 macrophage accumulation (CD206, IL-4, IL-10, and CX3CL1), and apoptotic/necrotic cell death in the obstructed kidneys of female mice but not male mice. However, DIZE treatment had no effect on ACE2 activity or mRNA expression.Conclusion: DIZE exacerbates UUO-induced renal fibrosis by aggravating tubular damage, apoptosis, and inflammation through independent of Ang (1–7), Ang 2 levels, and ACE2 expression/activity, rather than protecting against renal fibrosis after UUO. DIZE also has powerful effects on recruiting macrophages, including the M2-polarized subtype, in female UUO mice.
Collapse
Affiliation(s)
- Yosep Kim
- Department of Biotechnology, College of Fisheries Sciences, Pukyong National University, Busan, Republic of Korea
| | - Jongwan Kim
- Department of Medical Laboratory Science, Dong-Eui Institute of Technology, Busan, Republic of Korea
- Jongwan Kim Department of Medical Laboratory Science, Dong-Eui Institute of Technology, 54 Yangji-ro, Busanjin-gu, Busan 47230, Republic of Korea. E-mail:
| | - Sang Jun Han
- Department of Biotechnology, College of Fisheries Sciences, Pukyong National University, Busan, Republic of Korea
- Correspondence: Sang Jun Han Department of Biotechnology, College of Fisheries Sciences, Pukyong National University, 45 Yongso-ro, Nam-gu, Busan 48513, Republic of Korea. E-mail:
| |
Collapse
|
10
|
Fernandes RS, Netto MRT, Carvalho FB, Rigatto K. Alamandine: A promising treatment for fibrosis. Peptides 2022; 157:170848. [PMID: 35931236 DOI: 10.1016/j.peptides.2022.170848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 01/18/2023]
Abstract
Angiotensin (Ang) II, the main active member of the renin angiotensin system (RAS), is essential for the maintenance of cardiovascular homeostasis. However, hyperactivation of the RAS causes fibrotic diseases. Ang II has pro-inflammatory actions, and moreover activates interstitial fibroblasts and/or dysregulates extracellular matrix degradation. The discovery of new RAS pathways has revealed the complexity of this system. Among the RAS peptides, alamandine (ALA, Ala1 Ang 1-7) has been identified in humans, rats, and mice, with protective actions in different pathological conditions. ALA has similar effects to its well-known congener, Ang-(1-7), as a vasodilator, anti-inflammatory, and antifibrotic. Its protective role against cardiovascular diseases is well-reviewed in the literature. However, the protective actions of ALA in fibrotic conditions have been little explored. Therefore, in this article, we review the ability of ALA to modulate the inflammatory process and collagen deposition, to serve as an antioxidant, and to mediate protection against functional disorders. In this scenario, we also explore ALA as a promising therapy for pulmonary fibrosis after COVID-19 infection.
Collapse
Affiliation(s)
- Renata Streck Fernandes
- Laboratório de Fisiologia Translacional, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Brazil; Programa de Pós-graduação em Ciências da Saúde, UFCSPA, Brazil
| | | | | | - Katya Rigatto
- Laboratório de Fisiologia Translacional, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Brazil; Programa de Pós-graduação em Ciências da Saúde, UFCSPA, Brazil.
| |
Collapse
|
11
|
Haidar MA, Shakkour Z, Reslan MA, Al-Haj N, Chamoun P, Habashy K, Kaafarani H, Shahjouei S, Farran SH, Shaito A, Saba ES, Badran B, Sabra M, Kobeissy F, Bizri M. SARS-CoV-2 involvement in central nervous system tissue damage. Neural Regen Res 2022; 17:1228-1239. [PMID: 34782556 PMCID: PMC8643043 DOI: 10.4103/1673-5374.327323] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 06/30/2021] [Accepted: 07/28/2021] [Indexed: 12/18/2022] Open
Abstract
As the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to spread globally, it became evident that the SARS-CoV-2 virus infects multiple organs including the brain. Several clinical studies revealed that patients with COVID-19 infection experience an array of neurological signs ranging in severity from headaches to life-threatening strokes. Although the exact mechanism by which the SARS-CoV-2 virus directly impacts the brain is not fully understood, several theories have been suggested including direct and indirect pathways induced by the virus. One possible theory is the invasion of SARS-CoV-2 to the brain occurs either through the bloodstream or via the nerve endings which is considered to be the direct route. Such findings are based on studies reporting the presence of viral material in the cerebrospinal fluid and brain cells. Nevertheless, the indirect mechanisms, including blood-clotting abnormalities and prolonged activation of the immune system, can result in further tissue and organ damages seen during the course of the disease. This overview attempts to give a thorough insight into SARS-CoV-2 coronavirus neurological infection and highlights the possible mechanisms leading to the neurological manifestations observed in infected patients.
Collapse
Affiliation(s)
- Muhammad Ali Haidar
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Zaynab Shakkour
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mohammad Amine Reslan
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadine Al-Haj
- Faculty of Health Sciences, University of Balamand, Beirut, Lebanon
| | - Perla Chamoun
- Faculty of Medicine, University of Balamand, Koura, Lebanon
| | - Karl Habashy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Shima Shahjouei
- Program for Neurotrauma, Neuroproteomics & Biomarkers Research, Departments of Emergency Medicine, University of Florida, Gainesville, FL, USA
| | - Sarah H. Farran
- Department of Pathology and Laboratory Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | | | - Esber S. Saba
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Bassam Badran
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences I, Lebanese University, Hadath, Beirut, Lebanon
| | - Mirna Sabra
- Faculty of Medicine, Lebanese University, Neuroscience Research Center (NRC), Beirut, Lebanon
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Neuroscience Institute, Neurology Department, Geisinger Health System, PA, USA
| | - Maya Bizri
- Department of Psychiatry, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
12
|
Alimoradi N, Sharqi M, Firouzabadi D, Sadeghi MM, Moezzi MI, Firouzabadi N. SNPs of ACE1 (rs4343) and ACE2 (rs2285666) genes are linked to SARS-CoV-2 infection but not with the severity of disease. Virol J 2022; 19:48. [PMID: 35305693 PMCID: PMC8934128 DOI: 10.1186/s12985-022-01782-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/10/2022] [Indexed: 01/08/2023] Open
Abstract
COVID-19 and the renin-angiotensin system (RAS) are linked by angiotensin-converting enzyme 2 (ACE2), a key enzyme in RAS that has been validated as a SARS-CoV-2 receptor. Functional ACE1/ACE2 gene polymorphisms may lead to the imbalance between ACE/ACE2 ratio and thus generating RAS imbalance that is associated with higher degrees of lung damage in ARDS that may contribute to the COVID-19 infection outcome. Herein, we investigated the role of RAS gene polymorphisms, ACE1 (A2350G) and ACE2 (G8790A) as risk predictors for susceptibility and severity of COVID-19 infection. A total of 129 included: negative controls without a history of COVID-19 infection (n = 50), positive controls with a history of COVID-19 infection who were not hospitalized (n = 35), and patients with severe COVID-19 infection who were hospitalized in the intensive care unit (n = 44). rs4343 of ACE and rs2285666 of ACE2 were genotyped using PCR-RFLP method. Our results indicated that susceptibility to COVID-19 infection was associated with age, GG genotype of A2350G (Pa = 0.01; OR 4.7; 95% CI 1.4-15.1 and Pc = 0.040; OR 2.5; 95% CI 1.05-6.3) and GG genotype of G8790A (Pa = 0.044; OR 6.17; 95% CI 1.05-35.71 and Pc = 0.0001; OR 5.5; 95% CI 2.4-12.4). The G allele of A2350G (Pa = 0.21; OR 1.74; 95% CI 0.73-4.17 and Pc = 0.007; OR 2.1; 95% CI 1.2-3.5) and G allele of G8790A (Pa = 0.002; OR 4.26; 95% CI 1.7-10.65 and Pc = 0.0001; OR 4.7; 95% CI 2.4-9.2) were more frequent in ICU-admitted patients and positive control group. Also lung involvement due to COVID-19 infection was associated with age and the comorbidities such as diabetes. In conclusion, our findings support the association between the wild genotype (GG) of ACE2 and homozygote genotype (GG) of ACE1 and sensitivity to COVID-19 infection, but not its severity. However, confirmation of this hypothesis requires further studies with more participants.
Collapse
Affiliation(s)
- Nahid Alimoradi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Moein Sharqi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Dena Firouzabadi
- Shahid Faghihi Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
- Clinical Pharmacy Department, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Moein Sadeghi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Iman Moezzi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Firouzabadi
- Department of Pharmacology & Toxicology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
13
|
Elshafei A, Khidr EG, El-Husseiny AA, Gomaa MH. RAAS, ACE2 and COVID-19; a mechanistic review. Saudi J Biol Sci 2021; 28:6465-6470. [PMID: 34305426 PMCID: PMC8270731 DOI: 10.1016/j.sjbs.2021.07.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 05/26/2021] [Accepted: 07/04/2021] [Indexed: 01/08/2023] Open
Abstract
The use of angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) in coronavirus disease 2019 (COVID-19) patients has been claimed as associated with the risk of COVID-19 infection and its subsequent morbidities and mortalities. These claims were resulting from the possibility of upregulating the expression of angiotensin-converting enzyme 2 (ACE2), facilitation of SARS-CoV-2 entry, and increasing the susceptibility of infection in such treated cardiovascular patients. ACE2 and renin-angiotensin-aldosterone system (RAAS) products have a critical function in controlling the severity of lung injury, fibrosis, and failure following the initiation of the disease. This review is to clarify the mechanisms beyond the possible deleterious effects of angiotensin II (Ang II), and the potential protective role of angiotensin 1-7 (Ang 1-7) against pulmonary fibrosis, with a subsequent discussion of the latest updates on ACEIs/ARBs use and COVID-19 susceptibility in the light of these mechanisms and biochemical explanation.
Collapse
Key Words
- ACE1, angiotensin-converting enzyme 1
- ACE2
- ACE2, angiotensin-converting enzyme 2
- ACEIs
- ACEIs, angiotensin-converting enzyme inhibitors
- AEC-II, alveolar epithelial type II cells
- ARBs
- ARBs, angiotensin receptor blockers
- AT1R, angiotensin type 1 receptor
- AT2R, angiotensin type 2 receptor
- Ang 1-7, angiotensin 1-7
- Ang 1-9, angiotensin 1-9
- AngI, angiotensin I
- AngII, angiotensin II
- Angiotensin 1–7
- Angiotensin II
- COVID-19
- COVID-19, coronavirus disease 2019
- CVD, cardiovascular disease
- ERK, extracellular signal-regulated kinase
- ICU, intensive care unit
- MAPK, mitogen-activated protein kinase
- NLRP3, (NOD, LRR, and pyrin domain-containing protein 3)
- RAAS, renin-angiotensin-aldosterone system
- TGF-β, transforming growth factor-beta
- miR-21, microRNA-21
Collapse
Affiliation(s)
- Ahmed Elshafei
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Emad Gamil Khidr
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Ahmed A. El-Husseiny
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| | - Maher H. Gomaa
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy, Al-Azhar University, Nasr City 11231, Cairo, Egypt
| |
Collapse
|
14
|
Bastolla U. Mathematical Model of SARS-Cov-2 Propagation Versus ACE2 Fits COVID-19 Lethality Across Age and Sex and Predicts That of SARS. Front Mol Biosci 2021; 8:706122. [PMID: 34322518 PMCID: PMC8311794 DOI: 10.3389/fmolb.2021.706122] [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: 05/06/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
The fatality rate of Covid-19 escalates with age and is larger in men than women. I show that these variations correlate strongly with the level of the viral receptor protein ACE2 in rat lungs, which is consistent with the still limited data on human ACE2. Surprisingly, lower receptor levels correlate with higher fatality. I propose two possible explanations of this negative correlation: First, a previous mathematical model predicts that the velocity of viral progression in the organism as a function of the receptor level has a maximum and declines for abundant receptor. Secondly, degradation of ACE2 by the virus may cause the runaway inflammatory response that characterizes severe CoViD-19. I present here a mathematical model that predicts the lethality as a function of ACE2 protein level based on the two above hypothesis. The model fits Covid-19 fatality rate across age and sex in three countries with high accuracy (r 2 > 0.9 ) under the hypothesis that the speed of viral progression in the infected organism is a decreasing function of the ACE2 level. Moreover, rescaling the fitted parameters by the ratio of the binding rates of the spike proteins of SARS-CoV and SARS-CoV-2 allows predicting the fatality rate of SARS-CoV across age and sex, thus linking the molecular and epidemiological levels.
Collapse
Affiliation(s)
- Ugo Bastolla
- Centro de Biologia Molecular “Severo Ochoa”, CSIC-UAM Cantoblanco, Madrid, Spain
| |
Collapse
|
15
|
Garcia-Garduño TC, Padilla-Gutierrez JR, Cambrón-Mora D, Valle Y. RAAS: A Convergent Player in Ischemic Heart Failure and Cancer. Int J Mol Sci 2021; 22:7106. [PMID: 34281199 PMCID: PMC8268500 DOI: 10.3390/ijms22137106] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022] Open
Abstract
The current global prevalence of heart failure is estimated at 64.34 million cases, and it is expected to increase in the coming years, especially in countries with a medium-low sociodemographic index where the prevalence of risk factors is increasing alarmingly. Heart failure is associated with many comorbidities and among them, cancer has stood out as a contributor of death in these patients. This connection points out new challenges both in the context of the pathophysiological mechanisms involved, as well as in the quality of life of affected individuals. A hallmark of heart failure is chronic activation of the renin-angiotensin-aldosterone system, especially marked by a systemic increase in levels of angiotensin-II, a peptide with pleiotropic activities. Drugs that target the renin-angiotensin-aldosterone system have shown promising results both in the prevention of secondary cardiovascular events in myocardial infarction and heart failure, including a lower risk of certain cancers in these patients, as well as in current cancer therapies; therefore, understanding the mechanisms involved in this complex relationship will provide tools for a better diagnosis and treatment and to improve the prognosis and quality of life of people suffering from these two deadly diseases.
Collapse
Affiliation(s)
- Texali C. Garcia-Garduño
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara 44340, Mexico; (T.C.G.-G.); (J.R.P.-G.)
- Doctorado en Genética Humana, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Jorge R. Padilla-Gutierrez
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara 44340, Mexico; (T.C.G.-G.); (J.R.P.-G.)
| | - Diego Cambrón-Mora
- Doctorado en Biología Molecular, Departamento de Biología Molecular y Genómica, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Yeminia Valle
- Centro Universitario de Ciencias de la Salud, Instituto de Investigación en Ciencias Biomédicas, Universidad de Guadalajara, Guadalajara 44340, Mexico; (T.C.G.-G.); (J.R.P.-G.)
| |
Collapse
|
16
|
Uzunova VV, Todev A, Zarkos J, Addai D, Ananiev J, Rashev P, Alexandrova R, Tolekova A. Strengthening CoViD-19 therapy via combinations of RAS modulators. Med Hypotheses 2021; 150:110571. [PMID: 33799164 PMCID: PMC7992309 DOI: 10.1016/j.mehy.2021.110571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/09/2021] [Accepted: 03/13/2021] [Indexed: 02/07/2023]
Abstract
Evidence has accumulated that the pathology of CoViD-19 is strongly related to the renin-angiotensin system (RAS). The blockage of the angiotensin converting enzyme 2 (ACE2) by the SARS-CoV-2 virus leads to downstream consequences such as increased vascular tone, extensive fibrosis and pronounced immune reactions. Different approaches to tackle the adverse viral effects by compensating the lost ACE2 function have been suggested. Here, we use an unequal-arm lever model to describe a simplified version of the biased regulation exercised by the angiotensin II and angiotensin-(1-7) hormones, which are the substrate and the product of ACE2, respectively. We reason upon the lever dynamics and its disruptions caused by the virus, and propose that a combination of RAS modulators will most efficiently compensate the imbalance due to the excess of angiotensin II and the scarcity of angiotensin-(1-7). Specifically, we focus on the possible benefits of the simultaneous application of two agents, a MAS-receptor agonist and an angiotensin-II-type-2-receptor agonist. We conjecture that this combination has the potential to introduce a beneficial synergistic action that promotes anti-hypoxic, anti-fibrotic and anti-proliferative effects, thereby improving the clinical management of acute and chronic CoViD-19 pathologies.
Collapse
Affiliation(s)
| | - Angel Todev
- Trakia University, Medical Faculty, Department of Physiology, Pathophysiology and Pharmacology, Stara Zagora, Bulgaria
| | - Jacqueline Zarkos
- Trakia University, Medical Faculty, Department of Physiology, Pathophysiology and Pharmacology, Stara Zagora, Bulgaria
| | - Daniel Addai
- Trakia University, Medical Faculty, Department of Physiology, Pathophysiology and Pharmacology, Stara Zagora, Bulgaria
| | - Julian Ananiev
- Trakia University, Medical Faculty, Department of General and Clinical Pathology, Stara Zagora, Bulgaria
| | - Pavel Rashev
- Institute of Biology and Immunology of Reproduction "Acad. Kiril Bratanov", Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Radostina Alexandrova
- Institute of Experimental Morphology, Pathology and Anthropology Museum, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Anna Tolekova
- Trakia University, Medical Faculty, Department of Physiology, Pathophysiology and Pharmacology, Stara Zagora, Bulgaria; Trakia University, Medical College, Stara Zagora, Bulgaria.
| |
Collapse
|
17
|
Katsi V, Pavlidis G, Charalambous G, Tousoulis D, Toutouzas K. COVID-19, Angiotensin-Converting Enzyme 2 and Renin-Angiotensin System Inhibition: Implications for Practice. Curr Hypertens Rev 2021; 18:3-10. [PMID: 33475077 DOI: 10.2174/1573402117666210121100201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 11/16/2020] [Accepted: 11/16/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Recent studies suggested that patients with coronavirus disease 2019 (COVID-19) who use renin-angiotensin system (RAS) inhibitors have an increased risk of respiratory failure and death. The hypothesis was that angiotensin-converting enzyme inhibitor (ACEIs) or angiotensin receptor blocker (ARBs) may up-regulate ACE2 expression that is used as receptor for viral entry into cells. OBJECTIVE The purpose of this review is to discuss the existing evidence on the interaction between COVID-19 infection, ACE2 and ACEIs or ARBs and to examine the main implications for clinical practice. In addition, novel therapeutic strategies for blocking ACE2-mediated COVID-19 infection will be displayed. METHODS We performed a comprehensive review of the literature to identify data from clinical and experimental studies for the association between COVID-19 infection, ACE2 and RAS inhibition. RESULTS The current clinical and experimental evidence for ACEIs or ARBs to facilitate severe acute respiratory distress syndrome-coronavirus-2 (SARS-CoV-2) is insufficient to suggest discontinuing these drugs. Several observational studies arrive at the conclusion that the continued use of RAS inhibitors is unlike to be harmful in COVID-19-positive patients. CONCLUSIONS Further randomized trials are needed to answer definitely the question of whether RAS inhibitors are harmful or beneficial to patients with COVID-19.
Collapse
Affiliation(s)
- Vasiliki Katsi
- 1 st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens. Greece
| | - George Pavlidis
- Emergency Department, 'Hippokration' General Hospital, Athens. Greece
| | | | - Dimitrios Tousoulis
- 1 st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens. Greece
| | - Konstantinos Toutouzas
- 1 st Department of Cardiology, 'Hippokration' General Hospital, National and Kapodistrian University of Athens, School of Medicine, Athens. Greece
| |
Collapse
|
18
|
Carvalho PRD, Sirois P, Fernandes PD. The role of kallikrein-kinin and renin-angiotensin systems in COVID-19 infection. Peptides 2021; 135:170428. [PMID: 33065209 PMCID: PMC7553876 DOI: 10.1016/j.peptides.2020.170428] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/08/2020] [Accepted: 10/09/2020] [Indexed: 02/07/2023]
Abstract
In November 2019 the first cases of a novel acute respiratory syndrome has been reported in Wuhan province, China. Soon after, in January 2020 the World Health Organization declared a pandemic state due to the dissemination of a virus named SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), the cause of coronavirus disease 2019 (COVID-19). Being an unknown disease, it is essential to assess not only its main characteristic features and overall clinical symptomatology but also its patient infection mode and propagation to design appropriate clinical interventions and treatments. In this review the pathophysiology of SARS-CoV-2 infection and how the virus enters the cells and activates the immune system are described. The role of three systems involved in the SARS- CoV-2 infection (renin-angiotensin, kinin and coagulation systems) is discussed with the objectives to identify and try to explain several of the events observed during the evolution of the disease and to suggest possible targets for therapeutic interventions.
Collapse
Affiliation(s)
- Patricia Ribeiro de Carvalho
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Laboratório de Farmacologia da Dor e da Inflamação, Rio de Janeiro, Brazil
| | | | - Patricia Dias Fernandes
- Universidade Federal do Rio de Janeiro, Instituto de Ciências Biomédicas, Laboratório de Farmacologia da Dor e da Inflamação, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
ACE2, angiotensin 1-7 and skeletal muscle: review in the era of COVID-19. Clin Sci (Lond) 2020; 134:3047-3062. [PMID: 33231620 PMCID: PMC7687025 DOI: 10.1042/cs20200486] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/30/2020] [Accepted: 11/03/2020] [Indexed: 12/12/2022]
Abstract
Angiotensin converting enzyme-2 (ACE2) is a multifunctional transmembrane protein recently recognised as the entry receptor of the virus causing COVID-19. In the renin–angiotensin system (RAS), ACE2 cleaves angiotensin II (Ang II) into angiotensin 1-7 (Ang 1-7), which is considered to exert cellular responses to counteract the activation of the RAS primarily through a receptor, Mas, in multiple organs including skeletal muscle. Previous studies have provided abundant evidence suggesting that Ang 1-7 modulates multiple signalling pathways leading to protection from pathological muscle remodelling and muscle insulin resistance. In contrast, there is relatively little evidence to support the protective role of ACE2 in skeletal muscle. The potential contribution of endogenous ACE2 to the regulation of Ang 1-7-mediated protection of these muscle pathologies is discussed in this review. Recent studies have suggested that ACE2 protects against ageing-associated muscle wasting (sarcopenia) through its function to modulate molecules outside of the RAS. Thus, the potential association of sarcopenia with ACE2 and the associated molecules outside of RAS is also presented herein. Further, we introduce the transcriptional regulation of muscle ACE2 by drugs or exercise, and briefly discuss the potential role of ACE2 in the development of COVID-19.
Collapse
|
20
|
A potential protective role of losartan against coronavirus-induced lung damage. Infect Control Hosp Epidemiol 2020; 41:752-753. [PMID: 32248854 PMCID: PMC7137531 DOI: 10.1017/ice.2020.80] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
21
|
Namsolleck P, Moll GN. Does activation of the protective Renin-Angiotensin System have therapeutic potential in COVID-19? Mol Med 2020; 26:80. [PMID: 32807075 PMCID: PMC7430134 DOI: 10.1186/s10020-020-00211-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 08/11/2020] [Indexed: 12/20/2022] Open
Abstract
Infection of lung cells by the corona virus results in a loss of the balance between, on the one hand, angiotensin II-mediated stimulation of the angiotensin II type 1 receptor and, on the other hand, stimulation of the angiotensin II type 2 receptor and/or the Mas receptor. The unbalanced enhanced stimulation of the angiotensin II type 1 receptor causes inflammation, edema and contributes to the pathogenesis of severe acute respiratory distress syndrome. Here we hypothesize that stable, receptor-specific agonists of the angiotensin II type 2 receptor and of the Mas receptor are molecular medicines to treat COVID-19 patients. These agonists have therapeutic potential in the acute disease but in addition may reduce COVID-19-associated long-term pulmonary dysfunction and overall end-organ damage of this disease.
Collapse
Affiliation(s)
- Pawel Namsolleck
- Lanthio Pharma, a MorphoSys AG company, Rozenburglaan 13B, 9727 DL, Groningen, the Netherlands
| | - Gert N Moll
- Lanthio Pharma, a MorphoSys AG company, Rozenburglaan 13B, 9727 DL, Groningen, the Netherlands. .,Department of Molecular Genetics, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG, Groningen, the Netherlands.
| |
Collapse
|
22
|
Stancioiu F, Papadakis GZ, Kteniadakis S, Izotov BN, Coleman MD, Spandidos DA, Tsatsakis A. A dissection of SARS‑CoV2 with clinical implications (Review). Int J Mol Med 2020; 46:489-508. [PMID: 32626922 PMCID: PMC7307812 DOI: 10.3892/ijmm.2020.4636] [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: 05/15/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023] Open
Abstract
We are being confronted with the most consequential pandemic since the Spanish flu of 1918‑1920 to the extent that never before have 4 billion people quarantined simultaneously; to address this global challenge we bring to the forefront the options for medical treatment and summarize SARS‑CoV2 structure and functions, immune responses and known treatments. Based on literature and our own experience we propose new interventions, including the use of amiodarone, simvastatin, pioglitazone and curcumin. In mild infections (sore throat, cough) we advocate prompt local treatment for the naso‑pharynx (inhalations; aerosols; nebulizers); for moderate to severe infections we propose a tried‑and‑true treatment: the combination of arginine and ascorbate, administered orally or intravenously. The material is organized in three sections: i) Clinical aspects of COVID‑19; acute respiratory distress syndrome (ARDS); known treatments; ii) Structure and functions of SARS‑CoV2 and proposed antiviral drugs; iii) The combination of arginine‑ascorbate.
Collapse
Affiliation(s)
| | | | | | - Boris Nikovaevich Izotov
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119991 Moscow, Russia
| | - Michael D. Coleman
- School of Life and Health Sciences, Aston University, B4 7ET Birmingham, UK
| | | | - Aristidis Tsatsakis
- Department of Analytical and Forensic Medical Toxicology, Sechenov University, 119991 Moscow, Russia
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
23
|
AlQudah M, Hale TM, Czubryt MP. Targeting the renin-angiotensin-aldosterone system in fibrosis. Matrix Biol 2020; 91-92:92-108. [PMID: 32422329 DOI: 10.1016/j.matbio.2020.04.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 02/06/2023]
Abstract
Fibrosis is characterized by excessive deposition of extracellular matrix components such as collagen in tissues or organs. Fibrosis can develop in the heart, kidneys, liver, skin or any other body organ in response to injury or maladaptive reparative processes, reducing overall function and leading eventually to organ failure. A variety of cellular and molecular signaling mechanisms are involved in the pathogenesis of fibrosis. The renin-angiotensin-aldosterone system (RAAS) interacts with the potent Transforming Growth Factor β (TGFβ) pro-fibrotic pathway to mediate fibrosis in many cell and tissue types. RAAS consists of both classical and alternative pathways, which act to potentiate or antagonize fibrotic signaling mechanisms, respectively. This review provides an overview of recent literature describing the roles of RAAS in the pathogenesis of fibrosis, particularly in the liver, heart, kidney and skin, and with a focus on RAAS interactions with TGFβ signaling. Targeting RAAS to combat fibrosis represents a promising therapeutic approach, particularly given the lack of strategies for treating fibrosis as its own entity, thus animal and clinical studies to examine the impact of natural and synthetic substances to alter RAAS signaling as a means to treat fibrosis are reviewed as well.
Collapse
Affiliation(s)
- Mohammad AlQudah
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Canada; Department of Physiology and Biochemistry, College of Medicine, Jordan University of Science and Technology, Jordan
| | - Taben M Hale
- Department of Basic Medical Sciences, University of Arizona College of Medicine Phoenix, United States
| | - Michael P Czubryt
- Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, University of Manitoba, Canada.
| |
Collapse
|
24
|
D’Ardes D, Boccatonda A, Rossi I, Guagnano MT, Santilli F, Cipollone F, Bucci M. COVID-19 and RAS: Unravelling an Unclear Relationship. Int J Mol Sci 2020; 21:E3003. [PMID: 32344526 PMCID: PMC7215550 DOI: 10.3390/ijms21083003] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/31/2022] Open
Abstract
The renin-angiotensin system (RAS) plays a main role in regulating blood pressure and electrolyte and liquid balance. Previous evidence suggests that RAS may represent an important target for the treatment of lung pathologies, especially for acute respiratory distress syndrome and chronic fibrotic disease. The scientific community has recently focused its attention on angiotensin-converting enzyme (ACE) inhibitors and angiotensin receptor 1 (AT1R) inhibitors and their possible benefit/harms for patients infected by Coronavirus disease (COVID-19) who experience pneumonia, but there are still some doubts about the effects of these drugs in this setting.
Collapse
Affiliation(s)
| | | | | | | | - Francesca Santilli
- Clinica Medica Institute, European Center of Excellence on Atherosclerosis, Hypertension and Dyslipidemia, “G. D’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy (A.B.); (I.R.); (M.T.G.); (F.C.); (M.B.)
| | | | | |
Collapse
|
25
|
Association between preterm birth and the renin-angiotensin system in adolescence: influence of sex and obesity. J Hypertens 2019; 36:2092-2101. [PMID: 29846325 DOI: 10.1097/hjh.0000000000001801] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
OBJECTIVES Preterm birth appears to contribute to early development of cardiovascular disease, but the mechanisms are unknown. Prematurity may result in programming events that alter the renin-angiotensin system. We hypothesized that prematurity is associated with lower angiotensin-(1-7) in adolescence and that sex and obesity modify this relationship. METHODS We quantified angiotensin II and angiotensin-(1-7) in the plasma and urine of 175 adolescents born preterm and 51 term-born controls. We used generalized linear models to estimate the association between prematurity and the peptides, controlling for confounding factors and stratifying by sex and overweight/obesity. RESULTS Prematurity was associated with lower plasma angiotensin II (β: -5.2 pmol/l, 95% CI: -10.3 to -0.04) and angiotensin-(1-7) (-5.2 pmol/l, 95% CI: -8.4 to -2.0) but overall higher angiotensin II:angiotensin-(1-7) (3.0, 95% CI: 0.9-5.0). The preterm-term difference in plasma angiotensin-(1-7) was greater in women (-6.9 pmol/l, 95% CI: -10.7 to -3.1) and individuals with overweight/obesity (-8.0 pmol/l, 95% CI: -12.2 to -3.8). The preterm-term difference in angiotensin II:angiotensin-(1-7) was greater among those with overweight/obesity (4.4, 95% CI: 0.6-8.1). On multivariate analysis, prematurity was associated with lower urinary angiotensin II:angiotensin-(1-7) (-0.13, 95% CI: -0.26 to -0.003), especially among the overweight/obesity group (-0.38, 95% CI: -0.72 to -0.04). CONCLUSION Circulating angiotensin-(1-7) was diminished whereas urinary angiotensin-(1-7) was increased relative to angiotensin II in adolescents born preterm, suggesting prematurity may increase the risk of cardiovascular disease by altering the renin-angiotensin system. Perinatal renin-angiotensin system programming was more pronounced in women and individuals with overweight/obesity, thus potentially augmenting their risk of developing early cardiovascular disease.
Collapse
|
26
|
Chen Y, Zhao W, Liu C, Meng W, Zhao T, Bhattacharya SK, Sun Y. Molecular and Cellular Effect of Angiotensin 1-7 on Hypertensive Kidney Disease. Am J Hypertens 2019; 32:460-467. [PMID: 30715105 DOI: 10.1093/ajh/hpz009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/25/2018] [Accepted: 01/15/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Studies implicate that angiotensin 1-7 (Ang1-7) imparts protective effects in the kidney. However, its relevance in hypertensive kidney disease is not fully understood. The purpose of this study was to explore the role of Ang1-7 on renal damage/remodeling during hypertension and its potential underlying molecular-cellular mechanisms. METHODS Hypertension was induced in adult Sprague-Dawley rats by infusion of aldosterone (ALDO; 0.75 μg/hour) for 4 weeks with or without co-treatment of Ang1-7 (1 mg/kg/day). Untreated rats served as controls. Systolic blood pressure was monitored by tail-cuff technique. Renal fibrosis was evaluated by picrosirius red staining and renal collagen volume fraction was quantitated using imaging analyzing system. The expression of profibrotic factors [transforming growth factor-β1 (TGF-β1), platelet-derived growth factor-D (PDGF-D), fibroblast growth factor-1 (FGF-1), vascular endothelial growth factor-D (VEGF-D), and tissue inhibitors of metalloproteinases (TIMPs)] and free radical producing enzymes (inducible nitric oxide synthase and nicotinamide adenine dinucleotide phosphate [NADPH] oxidase) in the kidney were examined by reverse transcription-polymerase chain reaction and western blot. Renal oxidative stress was assessed by malondialdehyde (MDA) measurement. RESULTS Chronic ALDO infusion caused hypertension and hypertensive renal disease represented as glomerular damage/sclerosis. Ang1-7 co-treatment did not affect blood pressure in ALDO-treated rats, but significantly attenuated the glomerular damage/fibrosis. ALDO treatment significantly elevated renal expression of profibrogenic factors, including TGF-β1, TIMP-1/TIMP-2, FGF-1, PDGF-D, and VEGF-D, whereas Ang1-7 co-treatment significantly reduced renal TGF-β1, TIMP-1/TIMP-2, and FGF-1, but not PDGF-D and VEGF-D. Furthermore, ALDO infusion elevated NADPH oxidase (gp91phox) and MDA in the kidney, which was attenuated by Ang1-7 co-treatment. CONCLUSIONS Ang1-7 plays a protective role in the hypertensive kidney disease independent of blood pressure. The beneficial effects of Ang1-7 are likely mediated via suppressing TGF-β/FGF-1 pathways and oxidative stress.
Collapse
Affiliation(s)
- Yuanjian Chen
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Wenyuan Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Chang Liu
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Weixin Meng
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Tieqiang Zhao
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Syamal K Bhattacharya
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Yao Sun
- Division of Cardiovascular Diseases, Department of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| |
Collapse
|
27
|
South AM, Nixon PA, Chappell MC, Diz DI, Russell GB, Shaltout HA, O’Shea TM, Washburn LK. Obesity is Associated with Higher Blood Pressure and Higher Levels of Angiotensin II but Lower Angiotensin-(1-7) in Adolescents Born Preterm. J Pediatr 2019; 205:55-60.e1. [PMID: 30404738 PMCID: PMC6561332 DOI: 10.1016/j.jpeds.2018.09.058] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 08/21/2018] [Accepted: 09/20/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVES To evaluate if obesity is associated with increased angiotensin II (Ang II) and decreased angiotensin-(1-7) or Ang-(1-7) in the circulation and urine among adolescents born prematurely. STUDY DESIGN In a cross-sectional analysis of 175 14-year-olds born preterm with very low birth weight, we quantified plasma and urinary Ang II and Ang-(1-7) and compared their levels between subjects with overweight/obesity (body mass index ≥85th percentile, n = 61) and those with body mass index <85th percentile (n = 114) using generalized linear models, adjusted for race and antenatal corticosteroid exposure. RESULTS Overweight/obesity was associated with higher systolic blood pressure and a greater proportion with high blood pressure. After adjustment for confounders, overweight/obesity was associated with an elevated ratio of plasma Ang II to Ang-(1-7) (β: 0.57, 95% CI 0.23-0.91) and higher Ang II (β: 0.21 pmol/L, 95% CI 0.03-0.39) but lower Ang-(1-7) (β: -0.37 pmol/L, 95% CI -0.7 to -0.04). Overweight/obesity was associated with a higher ratio of urinary Ang II to Ang-(1-7) (β: 0.21, 95% CI -0.02 to 0.44), an effect that approached statistical significance. CONCLUSIONS Among preterm-born adolescents, overweight/obesity was associated with increased Ang II but reduced Ang-(1-7) in the circulation and the kidney as well as higher blood pressure. Obesity may compound the increased risk of hypertension and cardiovascular disease in individuals born prematurely by further augmenting the prematurity-associated imbalance in the renin-angiotensin system.
Collapse
Affiliation(s)
- Andrew M. South
- Department of Pediatrics, Wake Forest School of Medicine,Cardiovascular Sciences Center, Wake Forest School of Medicine,Division of Public Health Sciences, Department of Epidemiology and Prevention, Wake Forest School of Medicine
| | - Patricia A. Nixon
- Department of Pediatrics, Wake Forest School of Medicine,Department of Health and Exercise Science, Wake Forest University
| | - Mark C. Chappell
- Cardiovascular Sciences Center, Wake Forest School of Medicine,Department of Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine
| | - Debra I. Diz
- Cardiovascular Sciences Center, Wake Forest School of Medicine,Department of Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine
| | - Gregory B. Russell
- Division of Public Health Sciences, Department of Biostatistical Sciences, Wake Forest School of Medicine
| | - Hossam A. Shaltout
- Cardiovascular Sciences Center, Wake Forest School of Medicine,Department of Obstetrics and Gynecology, Wake Forest School of Medicine, Winston Salem, North Carolina, USA,Department of Pharmacology and Toxicology, School of Pharmacy, University of Alexandria, Egypt
| | - T. Michael O’Shea
- Department of Pediatrics, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Lisa K. Washburn
- Department of Pediatrics, Wake Forest School of Medicine,Cardiovascular Sciences Center, Wake Forest School of Medicine
| |
Collapse
|
28
|
Balance and circumstance: The renin angiotensin system in wound healing and fibrosis. Cell Signal 2018; 51:34-46. [PMID: 30071289 DOI: 10.1016/j.cellsig.2018.07.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/27/2018] [Accepted: 07/29/2018] [Indexed: 12/17/2022]
Abstract
The tissue renin angiotensin system (tRAS) is a locally-acting master-modulator of tissue homeostasis and regeneration. Through these abilities, it is emerging as an attractive target for therapies aiming to restore tissue homeostasis in conditions associated with disturbed wound healing. The tRAS can be divided into two axes - one being pro-inflammatory and pro-fibrotic and one being anti-inflammatory and anti-fibrotic. However, the division of the axes is fuzzy and imperfect as the axes are codependent and the outcome of tRAS activation is determined by the context. Although the tRAS is a local system it shares its key enzymes, ligands and receptors with the systemic RAS and is consequently also targeted by repurposing of drugs developed against the systemic RAS to manage hypertension. With a focus on the skin we will here discuss the tRAS, its involvement in physiological and pathological wound healing, and the therapeutic aptitude of its targeting to treat chronic wounds and fibrosis.
Collapse
|