1
|
Rukavina Mikusic NL, Silva MG, Erra Díaz FA, Pineda AM, Ferragut F, Gómez KA, Mazzitelli L, Gonzalez Maglio DH, Nuñez M, Santos RAS, Grecco HE, Gironacci MM. Alamandine, a protective component of the renin-angiotensin system, reduces cellular proliferation and interleukin-6 secretion in human macrophages through MasR-MrgDR heteromerization. Biochem Pharmacol 2024; 229:116480. [PMID: 39128587 DOI: 10.1016/j.bcp.2024.116480] [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: 06/05/2024] [Revised: 07/21/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Alamandine (ALA) exerts protective effects similar to angiotensin (Ang) (1-7) through Mas-related G protein-coupled receptor type D receptor (MrgDR) activation, distinct from Mas receptor (MasR). ALA induces anti-inflammatory effects in mice but its impact in human macrophages remains unclear. We aimed to investigate the anti-inflammatory effects of ALA in human macrophages. Interleukin (IL)-6 and IL-1β were measured by ELISA in human THP-1 macrophages and human monocyte-derived macrophages exposed to lipopolysaccharide (LPS). Consequences of MasR-MrgDR heteromerization were investigated in transfected HEK293T cells. ALA decreased IL-6 and IL-1β secretion in LPS-activated THP-1 macrophages. The ALA-induced decrease in IL-6 but not in IL-1β was prevented by MasR blockade and MasR downregulation, suggesting MasR-MrgDR interaction. In human monocyte-derived M1 macrophages, ALA decreased IL-1β secretion independently of MasR. MasR-MrgDR interaction was confirmed in THP-1 macrophages, human monocyte-derived macrophages, and transfected HEK293T cells. MasR and MrgDR formed a constitutive heteromer that was not influenced by ALA. ALA promoted Akt and ERK1/2 activation only in cells expressing MasR-MrgDR heteromers, and this effect was prevented by MasR blockade. While Ang-(1-7) reduced cellular proliferation in MasR -but not MrgDR- expressing cells, ALA antiproliferative effect was elicited in cells expressing MasR-MrgDR heteromers. ALA also induced an antiproliferative response in THP-1 cells and this effect was abolished by MasR blockade, reinforcing MasR-MrgDR interaction. MasR-MrgDR heteromerization is crucial for ALA-induced anti-inflammatory and antiproliferative responses in human macrophages. This study broaden our knowledge of the protective axis of the RAS, thus enabling novel therapeutic approaches in inflammatory-associated diseases.
Collapse
Affiliation(s)
- Natalia L Rukavina Mikusic
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | - Mauro G Silva
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | | | - Angélica M Pineda
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | - Fátima Ferragut
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (CONICET), Buenos Aires, Argentina
| | - Karina A Gómez
- Instituto de Investigaciones en Ingeniería Genética y Biología Molecular (CONICET), Buenos Aires, Argentina
| | - Luciana Mazzitelli
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | - Daniel H Gonzalez Maglio
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Inmunología-IDEHU, Buenos Aires, Argentina
| | - Myriam Nuñez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Cátedra de Matemáticas, Dpto de Físico-Matemáticas, Buenos Aires, Argentina
| | - Robson A S Santos
- Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Hernán E Grecco
- Universidad de Buenos Aires, Facultad de Ciencias Exactas y Naturales, Dpto. de Física, Buenos Aires, Argentina
| | - Mariela M Gironacci
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina.
| |
Collapse
|
2
|
Mendes EP, Ianzer D, Peruchetti DB, Santos RAS, Vieira MAR. Interaction of Angiotensin-(1-7) with kinins in the kidney circulation: Role of B 1 receptors. Peptides 2024; 179:171246. [PMID: 38821119 DOI: 10.1016/j.peptides.2024.171246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/19/2024] [Accepted: 05/22/2024] [Indexed: 06/02/2024]
Abstract
Changes in renal hemodynamics impact renal function during physiological and pathological conditions. In this context, renal vascular resistance (RVR) is regulated by components of the Renin-Angiotensin System (RAS) and the Kallikrein-Kinin System (KKS). However, the interaction between these vasoactive peptides on RVR is still poorly understood. Here, we studied the crosstalk between angiotensin-(1-7) and kinins on RVR. The right kidneys of Wistar rats were isolated and perfused in a closed-circuit system. The perfusion pressure and renal perfusate flow were continuously monitored. Ang-(1-7) (1.0-25.0 nM) caused a sustained, dose-dependent reduction of relative RVR (rRVR). This phenomenon was sensitive to 10 nM A-779, a specific Mas receptor (MasR) antagonist. Bradykinin (BK) promoted a sustained and transient reduction in rRVR at 1.25 nM and 125 nM, respectively. The transient effect was abolished by 4 μM des-Arg9-Leu8-bradykinin (DALBK), a specific kinin B1 receptor (B1R) antagonist. Accordingly, des-Arg9-bradykinin (DABK) 1 μM (a B1R agonist) increased rRVR. Interestingly, pre-perfusion of Ang-(1-7) changed the sustained reduction of rRVR triggered by 1.25 nM BK into a transient effect. On the other hand, pre-perfusion of Ang-(1-7) primed and potentiated the DABK response, this mechanism being sensitive to A-779 and DALBK. Binding studies performed with CHO cells stably transfected with MasR, B1R, and kinin B2 receptor (B2R) showed no direct interaction between Ang-(1-7) with B1R or B2R. In conclusion, our findings suggest that Ang-(1-7) differentially modulates kinin's effect on RVR in isolated rat kidneys. These results help to expand the current knowledge regarding the crosstalk between the RAS and KKS complex network in RVR.
Collapse
Affiliation(s)
| | - Danielle Ianzer
- Department of Physiological Sciences, ICB, UFG, Goiania, GO, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics, INCT-Nanobiofar, Belo Horizonte, MG, Brazil
| | - Diogo Barros Peruchetti
- Department of Physiology and Biophysics, ICB, UFMG, Belo Horizonte, MG, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics, INCT-Nanobiofar, Belo Horizonte, MG, Brazil
| | - Robson Augusto Souza Santos
- Department of Physiology and Biophysics, ICB, UFMG, Belo Horizonte, MG, Brazil; National Institute of Science and Technology in Nanobiopharmaceutics, INCT-Nanobiofar, Belo Horizonte, MG, Brazil
| | | |
Collapse
|
3
|
Khan MS, Shahid I, Greene SJ, Mentz RJ, DeVore AD, Butler J. Mechanisms of current therapeutic strategies for heart failure: more questions than answers? Cardiovasc Res 2023; 118:3467-3481. [PMID: 36536991 DOI: 10.1093/cvr/cvac187] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/19/2022] [Accepted: 10/21/2022] [Indexed: 12/24/2022] Open
Abstract
Heart failure (HF) is a complex, multifactorial and heterogeneous syndrome with substantial mortality and morbidity. Over the last few decades, numerous attempts have been made to develop targeted therapies that may attenuate the known pathophysiological pathways responsible for causing the progression of HF. However, therapies developed with this objective have sometimes failed to show benefit. The pathophysiological construct of HF with numerous aetiologies suggests that interventions with broad mechanisms of action which simultaneously target more than one pathway maybe more effective in improving the outcomes of patients with HF. Indeed, current therapeutics with clinical benefits in HF have targeted a wider range of intermediate phenotypes. Despite extensive scientific breakthroughs in HF research recently, questions persist regarding the ideal therapeutic targets which may help achieve maximum benefit. In this review, we evaluate the mechanism of action of current therapeutic strategies, the pathophysiological pathways they target and highlight remaining knowledge gaps regarding the mode of action of these interventions.
Collapse
Affiliation(s)
- Muhammad Shahzeb Khan
- From the Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
| | - Izza Shahid
- Division of Cardiovascular Prevention, Houston Methodist Academic Institute, Houston, TX, USA
| | - Stephen J Greene
- From the Division of Cardiology, Duke University School of Medicine, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - Robert J Mentz
- From the Division of Cardiology, Duke University School of Medicine, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - Adam D DeVore
- From the Division of Cardiology, Duke University School of Medicine, Durham, NC, USA.,Duke Clinical Research Institute, Durham, NC, USA
| | - Javed Butler
- Baylor Scott and White Research Institute, Baylor University Medical Center, 3434 Live Oak St Ste 501, Dallas 75204, TX, USA
| |
Collapse
|
4
|
Kate Gadanec L, Qaradakhi T, Renee McSweeney K, Matsoukas JM, Apostolopoulos V, Burrell LM, Zulli A. Diminazene aceturate uses different pathways to induce relaxation in healthy and atherogenic blood vessels. Biochem Pharmacol 2023; 208:115397. [PMID: 36566945 DOI: 10.1016/j.bcp.2022.115397] [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: 07/29/2022] [Revised: 12/17/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Diminazene aceturate (DIZE), a putative angiotensin-converting enzyme 2 (ACE2) activator, elicits relaxation in various animal models. This study aimed to determine the relaxing mechanisms in internal iliac arteries utilised by DIZE in healthy and atherogenic rabbit models. Studies were conducted on internal iliac artery rings retrieved from male New Zealand White rabbits fed a 4-week healthy control (n = 24) or atherogenic diet (n = 20). To investigate pathways utilised by DIZE to promote arterial relaxation, a DIZE dose response [10-9.0 M - 10-5.0 M] was performed on pre-contracted rings incubated with pharmaceuticals that target: components of the renin-angiotensin system; endothelial- and vascular smooth muscle-dependent mechanisms; protein kinases; and potassium channels. ACE2 expression was quantified by immunohistochemistry analysis following a 2 hr or 4 hr DIZE incubation. DIZE significantly enhanced vessel relaxation in atherogenic rings at doses [10-5.5 M] (p < 0.01) and [10-5.0 M] (p < 0.0001), when compared to healthy controls. Comprehensive results from functional isometric studies determined that DIZE causes relaxation via different mechanisms depending on pathology. For the first time, we report that in healthy blood vessels DIZE exerts its direct relaxing effect through ACE2/AT2R and NO/sGC pathways; however, in atherogenesis this switches to MasR, arachidonic acid pathway (i.e., COX1/2, EET and DHET), MCLP, Ca2+ activated voltage channels, AMPK and ERK1/2. Moreover, quantitative immunohistochemical analysis demonstrated that DIZE increases artery ACE2 expression in a time dependent manner. We provide a detailed investigation of DIZE's mechanisms and demonstrate for the first time that in healthy and atherogenic arteries DIZE provides beneficial effects through directly inducing relaxation, albeit via different pathways.
Collapse
Affiliation(s)
- Laura Kate Gadanec
- Institute for Health and Sport, Victoria University, Melbourne 3030, Victoria, Australia.
| | - Tawar Qaradakhi
- Institute for Health and Sport, Victoria University, Melbourne 3030, Victoria, Australia.
| | | | - John M Matsoukas
- Institute for Health and Sport, Victoria University, Melbourne 3030, Victoria, Australia; Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Alberta T2N 4N1, Canada; NewDrug PC, Patras Science Park, 26500 Patras, Greece.
| | - Vasso Apostolopoulos
- Institute for Health and Sport, Victoria University, Melbourne 3030, Victoria, Australia; Australian Institute for Musculoskeletal Science, Melbourne 3021, Victoria, Australia.
| | - Louise M Burrell
- Department of Medicine, Austin Health, University of Melbourne, Heidelberg 3084, Victoria, Australia.
| | - Anthony Zulli
- Institute for Health and Sport, Victoria University, Melbourne 3030, Victoria, Australia.
| |
Collapse
|
5
|
Xue X, Duan R, Zhang QQ, Wang SY, Gong PY, Yan E, Zhang YD, Jiang T. A non-peptidic MAS1 agonist AVE0991 alleviates hippocampal synaptic degeneration in rats with chronic cerebral hypoperfusion. Curr Neurovasc Res 2021; 18:343-350. [PMID: 34636310 DOI: 10.2174/1567202618666211012095210] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/10/2021] [Accepted: 07/14/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) is a contributing factor for neurodegenerative diseases. As a recently identified heptapeptide of the brain renin-angiotensin system, angiotensin-(1-7) was revealed to activate its receptor MAS1 and thus ameliorated cognitive impairments in rats with CCH. Since hippocampal synaptic degeneration represents an important pathological basis of cognitive deficits, we hypothesize that activation of MAS1-mediated signaling may alleviate CCH-induced synaptic degeneration in the hippocampus. METHODS In this study, we tested this hypothesis and uncovered the underlying mechanisms in a rat model of CCH induced by bilateral common carotid artery ligation surgery. At 1 week after the surgery, rats received a daily intraperitoneal injection of vehicle or a non-peptidic MAS1 agonist AVE0991 for 8 weeks. During this procedure, cerebral blood flow (CBF) was recorded. The levels of MAS1, amyloid-β (Aβ), neuroinflammatory cytokines, glial cell markers and synaptophysin in the hippocampus were assessed at the end of the treatment period. RESULTS We showed that AVE0991 significantly alleviated hippocampal synaptic degeneration in rats with CCH. This protection might be achieved by facilitating CBF recovery, reducing hippocampal Aβ levels and suppressing neuroinflammatory responses. CONCLUSIONS These findings indicate that MAS1-mediated signaling may represent a novel therapeutic target for CCH-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiao Xue
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006. China
| | - Rui Duan
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006. China
| | - Qiao-Quan Zhang
- Department of Pathology, Nanjing Brain Hospital, Nanjing Medical University, Nanjing, 210029. China
| | - Si-Yu Wang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006. China
| | - Peng-Yu Gong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006. China
| | - Yan E
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006. China
| | - Ying-Dong Zhang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006. China
| | - Teng Jiang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006. China
| |
Collapse
|
6
|
Alves DT, Mendes LF, Sampaio WO, Coimbra-Campos LMC, Vieira MAR, Ferreira AJ, Martins AS, Popova E, Todiras M, Qadri F, Alenina N, Bader M, Santos RAS, Campagnole-Santos MJ. Hemodynamic phenotyping of transgenic rats with ubiquitous expression of an angiotensin-(1-7)-producing fusion protein. Clin Sci (Lond) 2021; 135:2197-2216. [PMID: 34494083 DOI: 10.1042/cs20210599] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/17/2022]
Abstract
Activation of the angiotensin (Ang)-converting enzyme (ACE) 2/Ang-(1-7)/MAS receptor pathway of the renin-angiotensin system (RAS) induces protective mechanisms in different diseases. Herein, we describe the cardiovascular phenotype of a new transgenic rat line (TG7371) that expresses an Ang-(1-7)-producing fusion protein. The transgene-specific mRNA and the corresponding protein were shown to be present in all evaluated tissues of TG7371 with the highest expression in aorta and brain. Plasma Ang-(1-7) levels, measured by radioimmunoassay (RIA) were similar to control Sprague-Dawley (SD) rats, however high Ang-(1-7) levels were found in the hypothalamus. TG7371 showed lower baseline mean arterial pressure (MAP), assessed in conscious or anesthetized rats by telemetry or short-term recordings, associated with increased plasma atrial natriuretic peptide (ANP) and higher urinary sodium concentration. Moreover, evaluation of regional blood flow and hemodynamic parameters with fluorescent microspheres showed a significant increase in blood flow in different tissues (kidneys, mesentery, muscle, spleen, brown fat, heart and skin), with a resulting decrease in total peripheral resistance (TPR). TG7371 rats, on the other hand, also presented increased cardiac and global sympathetic tone, increased plasma vasopressin (AVP) levels and decreased free water clearance. Altogether, our data show that expression of an Ang-(1-7)-producing fusion protein induced a hypotensive phenotype due to widespread vasodilation and consequent fall in peripheral resistance. This phenotype was associated with an increase in ANP together with an increase in AVP and sympathetic drive, which did not fully compensate the lower blood pressure (BP). Here we present the hemodynamic impact of long-term increase in tissue expression of an Ang-(1-7)-fusion protein and provide a new tool to investigate this peptide in different pathophysiological conditions.
Collapse
Affiliation(s)
- Daniele T Alves
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
- Max-Delbrück Center for Molecular Medicine-MDC, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Beriln, Germany
| | - Luiz Felipe Mendes
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Walkyria O Sampaio
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leda M C Coimbra-Campos
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Aparecida R Vieira
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Anderson J Ferreira
- Department of Morphology, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Almir S Martins
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Elena Popova
- Max-Delbrück Center for Molecular Medicine-MDC, Berlin, Germany
| | - Mihail Todiras
- Max-Delbrück Center for Molecular Medicine-MDC, Berlin, Germany
| | | | - Natalia Alenina
- Max-Delbrück Center for Molecular Medicine-MDC, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Beriln, Germany
| | - Michael Bader
- Max-Delbrück Center for Molecular Medicine-MDC, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Beriln, Germany
- Institute for Biology, University of Lübeck, Lübeck, Germany
- Charité, University Medicine Berlin, Berlin, Germany
| | - Robson A S Santos
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria Jose Campagnole-Santos
- Department of Physiology and Biophysics and INCT-Nanobiopharmaceutics, ICB, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
7
|
Ahmad A, Raish M, Alkharfy KM. The potential role of thymoquinone in preventing the cardiovascular complications of COVID-19. Vascul Pharmacol 2021; 141:106899. [PMID: 34311073 PMCID: PMC8299308 DOI: 10.1016/j.vph.2021.106899] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/07/2021] [Accepted: 07/20/2021] [Indexed: 01/20/2023]
Abstract
A new virus strain detected in late 2019 and not previously described in humans is the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes corona virus disease (COVID-19). While potential therapeutic approaches for COVID-19 are being investigated, significant initiatives are being made to create protective drugs and study various antiviral agents to cure the infection. However, an effective treatment strategy against COVID-19 is worrisome inadequate. The objective of the present manuscript is to discuss the potential role of thymoquinone (TQ) in preventing the cardiovascular complications of COVID-19, focusing on viral inhibition, antioxidant potential, vascular effect, and cardiac protection. The multifunctional properties of TQ could potentially synergize with the activity of current therapeutic interventions and offer a basis for managing COVID-19 disease more effectively. Even though the experimental evidence is positive, a translational application of TQ in COVID-19 is timely warranted.
Collapse
Affiliation(s)
- Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid M Alkharfy
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
8
|
Abdel-Fattah MM, Elgendy ANAM, Mohamed WR. Xanthenone, ACE2 activator, counteracted gentamicin-induced nephrotoxicity in rats: Impact on oxidative stress and ACE2/Ang-(1-7) signaling. Life Sci 2021; 275:119387. [PMID: 33774027 DOI: 10.1016/j.lfs.2021.119387] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 03/15/2021] [Accepted: 03/23/2021] [Indexed: 11/29/2022]
Abstract
Nephrotoxicity is a rapid deterioration of kidney function due to exposure to nephrotoxic drugs as gentamicin. Gentamicin increases the generation of reactive oxygen species (ROS) leading to inflammatory responses and nuclear factor-κB (NF-κB) activation. The renal renin-angiotensin system (RAS) is considered a crucial regulator for physiological homeostasis and disease progression through the classic ACE/Ang-II/AT1 axis and its antagonist, ACE2/Ang-(1-7)/Mas axis which exerts an important role in the kidney. The present study evaluates the protective effects of the angiotensin-converting enzyme 2 (ACE2) activator; xanthenone; against experimental nephrotoxicity induced by gentamicin. Rats were divided into 4 groups, normal control, xanthenone (2 mg/kg, s.c), gentamicin (100 mg/kg, i.p. for one week) and xanthenone + gentamicin groups. Blood urea nitrogen (BUN) and serum creatinine levels were measured. The kidney tissues were used for estimating glutathione (GSH), superoxide dismutase (SOD), malondialdehyde (MDA), tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), NF-κB, Angiotensin II (AngII), and Ang-(1-7). In addition, histopathological examination and Western blot analysis of ACE2 expression were done. Xanthenone significantly restored serum levels of BUN and creatinine. Xanthenone exerted significant antioxidant effect as revealed by increased GSH content and SOD activity together with reduced MDA content. It exerted anti-inflammatory effect by significant reduction in TNF-α, NF-κB and IL-6 expression compared to gentamicin group. Xanthenone increased Ang-(1-7) and ACE2 expression while significantly decreased Ang-II expression. Histopathologically, xanthenone markedly counteracted gentamicin-induced renal aberrations. Activation of ACE2/Ang-(1-7) by xanthenone produced significant antioxidant and anti-inflammatory effects that counteracted gentamicin-induced nephrotoxicity.
Collapse
Affiliation(s)
- Maha M Abdel-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt.
| | - Abdel Nasser A M Elgendy
- Department of Pharmacology, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Wafaa R Mohamed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62514, Egypt
| |
Collapse
|
9
|
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
|
10
|
Suvorava T, Kaesemeyer W. Targeting the Vascular Endothelium in the Treatment of COVID-19. J Cardiovasc Pharmacol 2021; 77:1-3. [PMID: 33079828 DOI: 10.1097/fjc.0000000000000932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 09/30/2020] [Indexed: 12/21/2022]
Abstract
ABSTRACT The involvement of the vascular endothelium in the complications of coronavirus disease 2019 is now recognized. Chief among these are pulmonary endotheliitis, cytokine storm, endotoxic shock, and cardiovascular collapse. This Perspectives article is focused on therapeutical strategies to reduce the risk of these complications by targeting the vascular endothelium as a part of the overall treatment of coronavirus disease 2019.
Collapse
Affiliation(s)
- Tatsiana Suvorava
- Institute of Pharmacology and Clinical Pharmacology, University Hospital, Duesseldorf, Germany; and
| | | |
Collapse
|
11
|
Fetal Growth Restriction and Hypertension in the Offspring: Mechanistic Links and Therapeutic Directions. J Pediatr 2020; 224:115-123.e2. [PMID: 32450071 PMCID: PMC8086836 DOI: 10.1016/j.jpeds.2020.05.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 12/24/2022]
|
12
|
South AM, Brady TM, Flynn JT. ACE2 (Angiotensin-Converting Enzyme 2), COVID-19, and ACE Inhibitor and Ang II (Angiotensin II) Receptor Blocker Use During the Pandemic: The Pediatric Perspective. Hypertension 2020; 76:16-22. [PMID: 32367746 PMCID: PMC7289676 DOI: 10.1161/hypertensionaha.120.15291] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Potential but unconfirmed risk factors for coronavirus disease 2019 (COVID-19) in adults and children may include hypertension, cardiovascular disease, and chronic kidney disease, as well as the medications commonly prescribed for these conditions, ACE (angiotensin-converting enzyme) inhibitors, and Ang II (angiotensin II) receptor blockers. Coronavirus binding to ACE2 (angiotensin-converting enzyme 2), a crucial component of the renin-angiotensin-aldosterone system, underlies much of this concern. Children are uniquely impacted by the coronavirus, but the reasons are unclear. This review will highlight the relationship of COVID-19 with hypertension, use of ACE inhibitors and Ang II receptor blockers, and lifetime risk of cardiovascular disease from the pediatric perspective. We briefly summarize the renin-angiotensin-aldosterone system and comprehensively review the literature pertaining to the ACE 2/Ang-(1-7) pathway in children and the clinical evidence for how ACE inhibitors and Ang II receptor blockers affect this important pathway. Given the importance of the ACE 2/Ang-(1-7) pathway and the potential differences between adults and children, it is crucial that children are included in coronavirus-related research, as this may shed light on potential mechanisms for why children are at decreased risk of severe COVID-19.
Collapse
Affiliation(s)
- Andrew M. South
- Section of Nephrology, Department of Pediatrics, Wake Forest School of Medicine and Brenner Children’s Hospital
- Department of Surgery-Hypertension and Vascular Research, Wake Forest School of Medicine
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine
- Cardiovascular Sciences Center, Wake Forest School of Medicine, Winston Salem, NC
| | - Tammy M. Brady
- Division of Nephrology, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joseph T. Flynn
- Department of Pediatrics, University of Washington School of Medicine and Division of Nephrology, Seattle Children’s Hospital, Seattle, WA, USA
| |
Collapse
|
13
|
Macedo LM, de Ávila RI, Pedrino GR, Colugnati DB, Valadares MC, Lima EM, Borges CL, Kitten GT, Gava E, Castro CH. Effect of angiotensin II and angiotensin-(1-7) on proliferation of stem cells from human dental apical papilla. J Cell Physiol 2020; 236:366-378. [PMID: 32519379 DOI: 10.1002/jcp.29862] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 05/27/2020] [Accepted: 05/27/2020] [Indexed: 02/06/2023]
Abstract
The effects of the renin-angiotensin system (RAS) on stem cells isolated from human dental apical papilla (SCAPs) are completely unknown. Therefore, the aim of this study was to identify RAS components expressed in SCAPs and the effects of angiotensin (Ang) II and Ang-(1-7) on cell proliferation. SCAPs were collected from third molar teeth of adolescents and maintained in cell culture. Messenger RNA expression and protein levels of angiotensin-converting enzyme (ACE), ACE2, and Mas, Ang II type I (AT1) and type II (AT2) receptors were detected in SCAPs. Treatment with either Ang II or Ang-(1-7) increased the proliferation of SCAPs. These effects were inhibited by PD123319, an AT2 antagonist. While Ang II augmented mTOR phosphorylation, Ang-(1-7) induced ERK1/2 phosphorylation. In conclusion, SCAPs produce the main RAS components and both Ang II and Ang-(1-7) treatments induced cell proliferation mediated by AT2 activation through different intracellular mechanisms.
Collapse
Affiliation(s)
- Larissa M Macedo
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Renato I de Ávila
- Laboratory of Education and Research in In Vitro Toxicology (Tox In), Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Gustavo R Pedrino
- Department of Physiological Sciences, Centre for Neuroscience and Cardiovascular Research, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Diego B Colugnati
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Marize C Valadares
- Laboratory of Education and Research in In Vitro Toxicology (Tox In), Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Eliana M Lima
- Pharmaceutical Technology Laboratory, Faculty of Pharmacy, Federal University of Goiás, Goiânia, GO, Brazil
| | - Clayton L Borges
- Laboratory of Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Gregory T Kitten
- Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Elisandra Gava
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | - Carlos H Castro
- Integrative Laboratory of Cardiovascular and Neurological Pathophysiology, Department of Physiological Sciences, Institute of Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| |
Collapse
|
14
|
Mulvey JJ, Magro CM, Ma LX, Nuovo GJ, Baergen RN. WITHDRAWN: A mechanistic analysis placental intravascular thrombus formation in COVID-19 patients. Ann Diagn Pathol 2020; 46:151529. [PMID: 32361635 PMCID: PMC7195270 DOI: 10.1016/j.anndiagpath.2020.151529] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023]
Affiliation(s)
- J Justin Mulvey
- Department of Laboratory Medicine, Memorial Sloan-Kettering Cancer Center, United States of America
| | - Cynthia M Magro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, United States of America
| | - Lucy X Ma
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, United States of America
| | - Gerard J Nuovo
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States of America; Discovery Life Sciences, Powell, OH, United States of America
| | - Rebecca N Baergen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, United States of America
| |
Collapse
|
15
|
Mulvey JJ, Magro CM, Ma LX, Nuovo GJ, Baergen RN. Analysis of complement deposition and viral RNA in placentas of COVID-19 patients. Ann Diagn Pathol 2020; 46:151530. [PMID: 32387855 PMCID: PMC7182529 DOI: 10.1016/j.anndiagpath.2020.151530] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 01/08/2023]
Abstract
COVID-19, the disease caused by the novel Coronavirus, SARS-CoV-2, is increasingly being recognized as a systemic thrombotic and microvascular injury syndrome that may have its roots in complement activation. We had the opportunity to study the placental pathology of five full-term births to COVID-19 patients. All five exhibited histology indicative of fetal vascular malperfusion characterized by focal avascular villi and thrombi in larger fetal vessels. Vascular complement deposition in the placentas was not abnormal, and staining for viral RNA and viral spike protein was negative. While all cases resulted in healthy, term deliveries, these findings indicate the systemic nature of COVID-19 infection. The finding of vascular thrombosis without complement deposition may reflect the systemic nature of COVID-19's procoagulant effects unrelated to systemic complement activation. This paper explores thrombosis in the placentas COVID-19-positive patients at our hospital Potential prothrombotic mechanisms are explored. Direct infection of the placentas is ruled out as a cause.
Collapse
Affiliation(s)
- J Justin Mulvey
- Department of Laboratory Medicine, Memorial Sloan-Kettering Cancer Center, United States of America
| | - Cynthia M Magro
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, United States of America
| | - Lucy X Ma
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, United States of America
| | - Gerard J Nuovo
- The Ohio State University Comprehensive Cancer Center, Columbus, OH, United States of America; Discovery Life Sciences, Powell, OH, United States of America
| | - Rebecca N Baergen
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, United States of America.
| |
Collapse
|
16
|
Ademiluyi AO, Oyeniran OH, Oboh G. Tropical Food Spices: A Promising Panacea for the Novel Coronavirus Disease (COVID-19). EFOOD 2020. [DOI: 10.2991/efood.k.201022.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
17
|
Effects of Angiotensin-(1-7) and Angiotensin II on Acetylcholine-Induced Vascular Relaxation in Spontaneously Hypertensive Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6512485. [PMID: 31827689 PMCID: PMC6886389 DOI: 10.1155/2019/6512485] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/22/2019] [Accepted: 08/12/2019] [Indexed: 01/31/2023]
Abstract
Endothelial dysfunction of small arteries occurs in patients with hypertension and in various hypertensive models. Endothelial function is usually evaluated by the degree of acetylcholine- (ACh-) induced vascular relaxation. Our previous study has found that compared to Wistar-Kyoto rats (WKY), ACh-induced vasodilatation was attenuated significantly in the mesenteric artery (MA), coronary artery (CA), and pulmonary artery (PA) of spontaneously hypertensive rats (SHR). This study investigated the influence of angiotensin- (Ang-) (1-7) and Ang II on blood pressure and ACh-induced vascular relaxation, as well as their interactive roles and downstream signal pathways in SHR and WKY. Intravenous injection of Ang II significantly increased, while Ang-(1-7) decreased the mean arterial pressure (MAP) in SHR. Ang-(1-7) improved ACh-induced relaxation in the MA, CA, and PA of SHR, while Ang II further attenuated it, which were inhibited by pretreatment with Mas receptor antagonist A-779 or AT1 receptor antagonist losartan, respectively. Ang-(1-7) decreased the basal arterial tension, and Ang II induced great vasoconstriction in SHR. Pretreatment with Ang-(1-7) inhibited the Ang II-induced pressor response, vasoconstriction, and the effects on ACh-induced relaxation in SHR. AT1 receptor expression was higher, while nitric oxide (NO), cGMP, and protein kinase G (PKG) levels of arteries were lower in SHR than in WKY. Ang II decreased, while Ang-(1-7) increased, the levels of NO, cGMP, and PKG of arteries. In addition, pretreatment with Ang-(1-7) inhibited the Ang II-induced reduction of NO, cGMP, and PKG in SHR. These results indicate that the activation of the Mas receptor by Ang-(1-7) can improve endothelial function and decrease MAP in SHR and inhibit the deteriorative effect of Ang II on endothelial function through the NO-cGMP-PKG pathway.
Collapse
|
18
|
South AM, Shaltout HA, Washburn LK, Hendricks AS, Diz DI, Chappell MC. Fetal programming and the angiotensin-(1-7) axis: a review of the experimental and clinical data. Clin Sci (Lond) 2019; 133:55-74. [PMID: 30622158 PMCID: PMC6716381 DOI: 10.1042/cs20171550] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 11/20/2018] [Accepted: 12/03/2018] [Indexed: 02/07/2023]
Abstract
Hypertension is the primary risk factor for cardiovascular disease that constitutes a serious worldwide health concern and a significant healthcare burden. As the majority of hypertension has an unknown etiology, considerable research efforts in both experimental models and human cohorts has focused on the premise that alterations in the fetal and perinatal environment are key factors in the development of hypertension in children and adults. The exact mechanisms of how fetal programming events increase the risk of hypertension and cardiovascular disease are not fully elaborated; however, the focus on alterations in the biochemical components and functional aspects of the renin-angiotensin (Ang) system (RAS) has predominated, particularly activation of the Ang-converting enzyme (ACE)-Ang II-Ang type 1 receptor (AT1R) axis. The emerging view of alternative pathways within the RAS that may functionally antagonize the Ang II axis raise the possibility that programming events also target the non-classical components of the RAS as an additional mechanism contributing to the development and progression of hypertension. In the current review, we evaluate the potential role of the ACE2-Ang-(1-7)-Mas receptor (MasR) axis of the RAS in fetal programming events and cardiovascular and renal dysfunction. Specifically, the review examines the impact of fetal programming on the Ang-(1-7) axis within the circulation, kidney, and brain such that the loss of Ang-(1-7) expression or tone, contributes to the chronic dysregulation of blood pressure (BP) and cardiometabolic disease in the offspring, as well as the influence of sex on potential programming of this pathway.
Collapse
Affiliation(s)
- Andrew M South
- Department of Pediatrics, Section of Nephrology, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Department of Epidemiology and Prevention, Division of Public Health Sciences, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Cardiovascular Sciences Center, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Hypertension and Vascular Research, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
| | - Hossam A Shaltout
- Cardiovascular Sciences Center, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Department of Obstetrics and Gynecology, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Alexandria, Egypt
- Hypertension and Vascular Research, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Department of Surgery, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
| | - Lisa K Washburn
- Department of Pediatrics, Section of Nephrology, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Cardiovascular Sciences Center, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Hypertension and Vascular Research, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
| | - Alexa S Hendricks
- Cardiovascular Sciences Center, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Hypertension and Vascular Research, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
| | - Debra I Diz
- Cardiovascular Sciences Center, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Hypertension and Vascular Research, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Department of Surgery, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
| | - Mark C Chappell
- Cardiovascular Sciences Center, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A.
- Hypertension and Vascular Research, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
- Department of Surgery, Wake Forest School of Medicine, 526 Vine Street, Winston Salem, NC 27157, U.S.A
| |
Collapse
|
19
|
Casey S, Herath C, Rajapaksha I, Jones R, Angus P. Effects of angiotensin-(1-7) and angiotensin II on vascular tone in human cirrhotic splanchnic vessels. Peptides 2018; 108:25-33. [PMID: 30179652 DOI: 10.1016/j.peptides.2018.08.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 08/14/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022]
Abstract
Evidence suggests that the renin angiotensin system (RAS) may play a role in the pathological splanchnic vasodilatation that leads to a hyperdynamic circulation in cirrhosis. An impaired contractile response to the angiotensin II peptide of the classical RAS system has been described in animal models of cirrhosis and in vivo in cirrhotic subjects. Furthermore, in experimental cirrhosis, the so-called alternate arm of the RAS was found to be upregulated and its effector peptide, angiotensin-(1-7) was shown to attenuate splanchnic vascular tone. The aim of this study was to explore the relevance of these findings to human disease. Omental arteries from cirrhotic and controls subjects were studied in isolation using a wire myograph. Varied protocols to evaluate the vasoactivity of RAS mediators were enacted. The contractile response to angiotensin II was comparable in cirrhotic vs control splanchnic arteries (61 ± 9 vs 68 ± 11% KPSS, respectively). Despite this, however, arterial contractility of the cirrhotic vessels correlated negatively with Child Pugh score (p = 0.0003, r=-0.83) and there was evidence that angiotensin II-induced contractility was increased in early cirrhosis. Angiotensin II-induced contractility was attenuated by angiotensin-(1-7) in cirrhotic and control arteries, however, adrenergic responses were not affected by angiotensin-(1-7). Contractile responses to angiotensin II are preserved in narrow lumen human cirrhotic splanchnic arteries and are comparatively augmented in early disease. Angiotensin-(1-7) had no vasodilatory effect on adrenergic tone, however, attenuated angiotensin II-induced contractility, possibly through an Ang-(1-7)-AT1R interaction, and thus may contribute to pathological vasodilatation in human cirrhosis.
Collapse
Affiliation(s)
- Stephen Casey
- Liver Unit, Austin Health, Melbourne, Australia; Department of Medicine, Austin Health, University of Melbourne, Australia.
| | - Chandana Herath
- Department of Medicine, Austin Health, University of Melbourne, Australia
| | - Indu Rajapaksha
- Department of Medicine, Austin Health, University of Melbourne, Australia
| | | | - Peter Angus
- Liver Unit, Austin Health, Melbourne, Australia; Department of Medicine, Austin Health, University of Melbourne, Australia
| |
Collapse
|
20
|
Chappell MC. Therapeutic Approaches to the Alternative Angiotensin-(1-7) Axis of the Renin-Angiotensin System. ANNALS OF PHARMACOLOGY AND PHARMACEUTICS 2017; 2:1116. [PMID: 36643780 PMCID: PMC9836034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Cardiovascular disease remains the leading cause of death for both men and women in the United States despite the recent advances in drug development, changes in lifestyle and screening protocols. A key target in the treatment of cardiovascular disease and hypertension is the renin-angiotensinsystem (RAS), a circulating and tissue system involved in the regulation of blood pressure, fluid balance and cellular injury. Pharmacologic approaches have traditionally focused on the Ang II-AT1receptor axis of the RAS to prevent the generation of Ang II with angiotensin converting enzyme inhibitors (ACEI) or to block the binding of Ang II to the AT1 receptor (AT1R) with selective antagonists (ARBs).
Collapse
Affiliation(s)
- Mark C Chappell
- Correspondence: Mark C Chappell, Department of Surgery, Hypertensin and Vascular Research, Cardiovascular Sciences Center, Wake Forest School of Medicine, USA, Tel: (336) 716-9236; Fax: (336) 716-2456;
| |
Collapse
|
21
|
Hamias R, Rudich A, Greenberg G, Szendro G, Wolak T. Angiotensin 1–7, but not the thrombin-cleaved osteopontin C-terminal fragment, attenuates osteopontin-mediated macrophage-induced endothelial-cell inflammation. Inflamm Res 2017; 67:265-275. [DOI: 10.1007/s00011-017-1120-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 11/11/2017] [Accepted: 11/22/2017] [Indexed: 12/22/2022] Open
|
22
|
Chappell MC, Al Zayadneh EM. Angiotensin-(1-7) and the Regulation of Anti-Fibrotic Signaling Pathways. ACTA ACUST UNITED AC 2017; 2. [PMID: 28691117 DOI: 10.4172/2576-1471.1000134] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Mark C Chappell
- Department of Surgery/Hypertension and Vascular Research, Cardiovascular Sciences Center, Wake Forest School of Medicine Winston-Salem, NC, USA
| | - Ebaa M Al Zayadneh
- Department of Physiology and Biochemistry, University of Jordan, Amman, Jordon
| |
Collapse
|