1
|
Wu Q, Gille C, Maderspacher F, Hildebrand B, Thienel M, Clauss S. Angiotensin IV does not exert prothrombotic effects in vivo. JOURNAL OF MOLECULAR AND CELLULAR CARDIOLOGY PLUS 2025; 11:100287. [PMID: 40028176 PMCID: PMC11871494 DOI: 10.1016/j.jmccpl.2025.100287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/03/2025] [Accepted: 02/10/2025] [Indexed: 03/05/2025]
Abstract
Thrombosis and thromboembolism are serious clinical complications of cardiovascular diseases and are among the leading causes of mortality worldwide. Dysregulation of the renin-angiotensin system is associated with an increased incidence of thrombotic events. Angiotensin II (AngII) is known to enhance platelet aggregation, contributing to a prothrombotic state in patients. Important biological roles of other angiotensin peptides and their receptors have been shown, but their specific role in thrombus formation remains unclear. Recent evidence suggests a prothrombotic role of angiotensin IV (AngIV). To confirm the prothrombotic effects of AngIV and to further investigate AngIV-mediated mechanisms, we utilized osmotic minipumps to administer AngIV in mice continuously over 4 weeks. AngIV treatment did not induce thrombus formation in the heart, did not affect platelet numbers, and did not enhance platelet aggregation. HGF, c-MET, or PAI-1 expression levels in the heart were not affected by AngIV treatment in mice. Furthermore, we did not observe altered platelet aggregation of human platelets incubated with HGF. These findings indicate that AngIV does not regulate key prothrombotic mechanisms.
Collapse
Affiliation(s)
- Qifang Wu
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Christine Gille
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Florian Maderspacher
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
| | - Bianca Hildebrand
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Manuela Thienel
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| | - Sebastian Clauss
- Department of Medicine I, University Hospital, LMU Munich, Munich, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Munich, Munich Heart Alliance (MHA), Munich, Germany
- Institute of Surgical Research at the Walter-Brendel-Centre of Experimental Medicine, University Hospital, LMU Munich, Munich, Germany
- Interfaculty Center for Endocrine and Cardiovascular Disease Network Modelling and Clinical Transfer (ICONLMU), LMU Munich, Munich, Germany
| |
Collapse
|
2
|
Heath SP, Hermanns VC, Coucha M, Abdelsaid M. SARS-CoV-2 Spike Protein Exacerbates Thromboembolic Cerebrovascular Complications in Humanized ACE2 Mouse Model. Transl Stroke Res 2024:10.1007/s12975-024-01301-5. [PMID: 39354270 PMCID: PMC11973837 DOI: 10.1007/s12975-024-01301-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 08/09/2024] [Accepted: 09/26/2024] [Indexed: 10/03/2024]
Abstract
COVID-19 increases the risk for acute ischemic stroke, yet the molecular mechanisms are unclear and remain unresolved medical challenges. We hypothesize that the SARS-CoV-2 spike protein exacerbates stroke and cerebrovascular complications by increasing coagulation and decreasing fibrinolysis by disrupting the renin-angiotensin-aldosterone system (RAAS). A thromboembolic model was induced in humanized ACE2 knock-in mice after one week of SARS-CoV-2 spike protein injection. hACE2 mice were treated with Losartan, an angiotensin receptor (AT1R) blocker, immediately after spike protein injection. Cerebral blood flow and infarct size were compared between groups. Vascular-contributes to cognitive impairments and dementia was assessed using a Novel object recognition test. Tissue factor-III and plasminogen activator inhibitor-1 were measured using immunoblotting to assess coagulation and fibrinolysis. Human brain microvascular endothelial cells (HBMEC) were exposed to hypoxia with/without SARS-CoV-2 spike protein to mimic ischemic conditions and assessed for inflammation, RAAS balance, coagulation, and fibrinolysis. Our results showed that the SARS-CoV-2 spike protein caused an imbalance in the RAAS that increased the inflammatory signal and decreased the RAAS protective arm. SARS-CoV-2 spike protein increased coagulation and decreased fibrinolysis when coincident with ischemic insult, which was accompanied by a decrease in cerebral blood flow, an increase in neuronal death, and a decline in cognitive function. Losartan treatment restored RAAS balance and reduced spike protein-induced effects. SARS-CoV-2 spike protein exacerbates inflammation and hypercoagulation, leading to increased neurovascular damage and cognitive dysfunction. However, the AT1R blocker, Losartan, restored the RAAS balance and reduced COVID-19-induced thromboembolic cerebrovascular complications.
Collapse
Affiliation(s)
- Stan P Heath
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Veronica C Hermanns
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, GA, USA
| | - Mohammed Abdelsaid
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA.
- Biomedical Sciences Department, Mercer University School of Medicine, 1250 E 66th Street | Savannah, 31404, Macon, GA, United States.
| |
Collapse
|
3
|
Furon J, Yetim M, Pouettre E, Martinez de Lizarrondo S, Maubert E, Hommet Y, Lebouvier L, Zheng Z, Ali C, Vivien D. Blood tissue Plasminogen Activator (tPA) of liver origin contributes to neurovascular coupling involving brain endothelial N-Methyl-D-Aspartate (NMDA) receptors. Fluids Barriers CNS 2023; 20:11. [PMID: 36737775 PMCID: PMC9896721 DOI: 10.1186/s12987-023-00411-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Regulation of cerebral blood flow (CBF) directly influence brain functions and dysfunctions and involves complex mechanisms, including neurovascular coupling (NVC). It was suggested that the serine protease tissue-type plasminogen activator (tPA) could control CNV induced by whisker stimulation in rodents, through its action on N-methyl-D-Aspartate receptors (NMDARs). However, the origin of tPA and the location and mechanism of its action on NMDARs in relation to CNV remained debated. METHODS Here, we answered these issues using tPANull mice, conditional deletions of either endothelial tPA (VECad-CreΔtPA) or endothelial GluN1 subunit of NMDARs (VECad-CreΔGluN1), parabioses between wild-type and tPANull mice, hydrodynamic transfection-induced deletion of liver tPA, hepatectomy and pharmacological approaches. RESULTS We thus demonstrate that physiological concentrations of vascular tPA, achieved by the bradykinin type 2 receptors-dependent production and release of tPA from liver endothelial cells, promote NVC, through a mechanism dependent on brain endothelial NMDARs. CONCLUSIONS These data highlight a new mechanism of regulation of NVC involving both endothelial tPA and NMDARs.
Collapse
Affiliation(s)
- Jonathane Furon
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Mervé Yetim
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Elsa Pouettre
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Sara Martinez de Lizarrondo
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Eric Maubert
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Yannick Hommet
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Laurent Lebouvier
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Ze Zheng
- grid.30760.320000 0001 2111 8460Department of Medicine, Medical College of Wisconsin, Milwaukee, WI USA ,grid.280427.b0000 0004 0434 015XBlood Research Institute, Versiti Blood Center of Wisconsin, Milwaukee, WI USA
| | - Carine Ali
- grid.460771.30000 0004 1785 9671UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074 Caen, France
| | - Denis Vivien
- UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Normandie University, Bvd Becquerel, BP 5229, 14074, Caen, France. .,Department of Clinical Research, Caen-Normandie University Hospital, Caen, France.
| |
Collapse
|
4
|
Hossein BR, Kheirollah A, Seif F. Endothelin-1 Stimulates PAI-1 Protein Expression via Dual Transactivation Pathway Dependent ROCK and Phosphorylation of Smad2L. CELL JOURNAL 2022; 24:465-472. [PMID: 36093806 PMCID: PMC9468725 DOI: 10.22074/cellj.2022.7720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Indexed: 11/04/2022]
Abstract
<strong>Objective:</strong> In addition to the carboxy region, Smad2 transcription factor can be phosphorylated in the linker region as<br />well. Phosphorylation of Smad2 linker region (Smad2L) promotes the expression of plasminogen activator inhibitor type<br />1 (PAI-1) which leads to cardiovascular disorders such as atherosclerosis. The purpose of this study was to evaluate the role of dual transactivation of EGF and TGF-β receptors in phosphorylation of Smad2L and protein expression of PAI-1 induced by endothelin-1 (ET-1) in bovine aortic endothelial cells (BAECs). In addition, as an intermediary of G protein-coupled receptor (GPCR) signaling, the functions of ROCK and PLC were investigated in dual transactivation pathways.<br /><strong>Materials and Methods:</strong> The experimental study is an in vitro study performed on BAECs. Proteins were investigated<br />by western blotting using protein-specific antibodies against phospho-Smad2 linker region residues (Ser245/250/255),<br />phospho-Smad2 carboxy residues (465/467), ERK1/(Thr202/Thr204), and PAI-1.<br /><strong>Results:</strong> TGF (2 ng/ml), EGF (100 ng/ml) and ET-1 (100 nM) induced the phosphorylation of Smad2L. This response was<br />blocked in the presence of AG1478 (EGFR antagonists), SB431542 (TGFR inhibitor), and Y27632 (Rho-associated protein kinase (ROCK antagonist). Moreover, ET-1-increased protein expression of PAI-1 was decreased in the presence of bosentan (ET receptor inhibitor), AG1478, SB431542, and Y27632.<br /><strong>Conclusion:</strong> The results indicated that ET-1 increases the phosphorylation of Smad2L and protein expression of PAI-1<br />via induced the transactivation pathways of EGFR and TGFR. This study is the first attempt to scrutinize the significant role of ROCK in the protein expression of PAI-1.
Collapse
Affiliation(s)
- Babaahmadi-Rezaei Hossein
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical
Sciences, Ahvaz, Iran
| | - Alireza Kheirollah
- Department of Biochemistry, Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Faezeh Seif
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical
Sciences, Ahvaz, Iran,Department of Basic Sciences, Shoushtar Faculty of Medical Sciences, Shoushtar, Iran ,P.O.Box: 159Hyperlipidemia Research CenterDepartment of Clinical BiochemistryFaculty of MedicineAhvaz Jundishapur University of Medical SciencesAhvazIran
| |
Collapse
|
5
|
Angiotensin-Converting Enzyme (ACE) 1 Gene Polymorphism and Phenotypic Expression of COVID-19 Symptoms. Genes (Basel) 2021; 12:genes12101572. [PMID: 34680966 PMCID: PMC8535484 DOI: 10.3390/genes12101572] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/16/2021] [Accepted: 09/28/2021] [Indexed: 12/25/2022] Open
Abstract
The renin–angiotensin–aldosterone system (RAAS) appears to play an important role in SARS-CoV-2 infection. Polymorphisms within the genes that control this enzymatic system are candidates for elucidating the pathogenesis of COVID-19, since COVID-19 is not only a pulmonary disease but also affects many organs and systems throughout the body in multiple ways. Most striking is the fact that ACE2, one of the major components of the RAAS, is a prerequisite for SARS-COV-2 infection. Recently, we and other groups reported an association between a polymorphism of the ACE1 gene (a homolog of ACE2) and the phenotypic expression of COVID-19, particularly in its severity. The ethnic difference in ACE1 insertion (I)/deletion (D) polymorphism seems to explain the apparent difference in mortality between the West and East Asia. The purpose of this review was to further evaluate the evidence linking ACE1 polymorphisms to COVID-19. We searched the Medline database (2019–2021) for reference citations of relevant articles and selected studies on the clinical outcome of COVID-19 related to ACE1 I/D polymorphism. Although the numbers of patients are not large enough yet, most available evidence supports the notion that the DD genotype adversely influences COVID-19 symptoms. Surprisingly, small studies conducted in several countries yielded opposite results, suggesting that the ACE1 II genotype is a risk factor. This contradictory result may be the case in certain geographic areas, especially in subgroups of patients. It may also be due to interactions with other genes or to yet unexplained biochemical mechanisms. According to our hypothesis, such candidates are genes that are functionally involved in the pathophysiology of COVID-19, can act in concert with the ACE1 DD genotype, and that show differences in their frequency between the West and East Asia. For this, we conducted research focusing on Alu-related genes. The current study on the ACE1 genotype will provide potentially new clues to the pathogenesis, treatment, and diagnosis of SARS-CoV-2 infections.
Collapse
|
6
|
Ekholm M, Kahan T. The Impact of the Renin-Angiotensin-Aldosterone System on Inflammation, Coagulation, and Atherothrombotic Complications, and to Aggravated COVID-19. Front Pharmacol 2021; 12:640185. [PMID: 34220496 PMCID: PMC8245685 DOI: 10.3389/fphar.2021.640185] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 06/07/2021] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is considered a disease caused by a chronic inflammation, associated with endothelial dysfunction, and several mediators of inflammation are up-regulated in subjects with atherosclerotic disease. Healthy, intact endothelium exhibits an antithrombotic, protective surface between the vascular lumen and vascular smooth muscle cells in the vessel wall. Oxidative stress is an imbalance between anti- and prooxidants, with a subsequent increase of reactive oxygen species, leading to tissue damage. The renin-angiotensin-aldosterone system is of vital importance in the pathobiology of vascular disease. Convincing data indicate that angiotensin II accelerates hypertension and augments the production of reactive oxygen species. This leads to the generation of a proinflammatory phenotype in human endothelial and vascular smooth muscle cells by the up-regulation of adhesion molecules, chemokines and cytokines. In addition, angiotensin II also seems to increase thrombin generation, possibly via a direct impact on tissue factor. However, the mechanism of cross-talk between inflammation and haemostasis can also contribute to prothrombotic states in inflammatory environments. Thus, blocking of the renin-angiotensin-aldosterone system might be an approach to reduce both inflammatory and thrombotic complications in high-risk patients. During COVID-19, the renin-angiotensin-aldosterone system may be activated. The levels of angiotensin II could contribute to the ongoing inflammation, which might result in a cytokine storm, a complication that significantly impairs prognosis. At the outbreak of COVID-19 concerns were raised about the use of angiotensin converting enzyme inhibitors and angiotensin receptor blocker drugs in patients with COVID-19 and hypertension or other cardiovascular comorbidities. However, the present evidence is in favor of continuing to use of these drugs. Based on experimental evidence, blocking the renin-angiotensin-aldosterone system might even exert a potentially protective influence in the setting of COVID-19.
Collapse
Affiliation(s)
- M Ekholm
- Karolinska Institutet, Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Stockholm, Sweden
| | - T Kahan
- Karolinska Institutet, Department of Clinical Sciences, Danderyd Hospital, Division of Cardiovascular Medicine, Stockholm, Sweden
| |
Collapse
|
7
|
Li M, Schifanella L, Larsen PA. Alu retrotransposons and COVID-19 susceptibility and morbidity. Hum Genomics 2021; 15:2. [PMID: 33390179 PMCID: PMC7779329 DOI: 10.1186/s40246-020-00299-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 12/14/2020] [Indexed: 12/22/2022] Open
Abstract
SARS-CoV-2 has spread rapidly across the world and is negatively impacting the global human population. COVID-19 patients display a wide variety of symptoms and clinical outcomes, including those attributed to genetic ancestry. Alu retrotransposons have played an important role in human evolution, and their variants influence host response to viral infection. Intronic Alus regulate gene expression through several mechanisms, including both genetic and epigenetic pathways. With respect to SARS-CoV-2, an intronic Alu within the ACE gene is hypothesized to be associated with COVID-19 susceptibility and morbidity. Here, we review specific Alu polymorphisms that are of particular interest when considering host response to SARS-CoV-2 infection, especially polymorphic Alu insertions in genes associated with immune response and coagulation/fibrinolysis cascade. We posit that additional research focused on Alu-related pathways could yield novel biomarkers capable of predicting clinical outcomes as well as patient-specific treatment strategies for COVID-19 and related infectious diseases.
Collapse
Affiliation(s)
- Manci Li
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, 55108, USA
| | - Luca Schifanella
- Department of Surgery, Division of Surgical Outcomes and Precision Medicine Research, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Peter A Larsen
- Department of Veterinary and Biomedical Sciences, University of Minnesota, St. Paul, MN, 55108, USA.
| |
Collapse
|
8
|
Zadeh Fakhar HB, Zali H, Rezaie-Tavirani M, Darkhaneh RF, Babaabasi B. Proteome profiling of low grade serous ovarian cancer. J Ovarian Res 2019; 12:64. [PMID: 31315664 PMCID: PMC6637464 DOI: 10.1186/s13048-019-0535-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/28/2019] [Indexed: 12/20/2022] Open
Abstract
Background Serous carcinoma, the subtype of ovarian cancer has the highest occurrence and mortality in women. Proteomic profiling using mass spectrometry (MS) has been used to detect biomarkers in tissue s obtained from patients with ovarian cancer. Thus, this study aimed at analyzing the interactome (protein-protein interaction (PPI)) and (MS) data to inspect PPI networks in patients with Low grade serous ovarian cancer. Methods For proteome profiling in Low grade serous ovarian cancer, 2DE and mass spectrometry were used. Differentially expressed proteins which had been determined in Low grade serous ovarian cancer and experimental group separately were integrated with PPI data to construct the (QQPPI) networks. Results Six Hub-bottlenecks proteins with significant centrality values, based on centrality parameters of the network (Degree and between), were found including Transgelin (TAGLN), Keratin (KRT14), Single peptide match to actin, cytoplasmic 1(ACTB), apolipoprotein A-I (APOA1), Peroxiredoxin-2 (PRDX2), and Haptoglobin (HP). Discussion This study showed these six proteins were introduced as hub-bottleneck protein. It can be concluded that regulation of gene expression can have a critical role in the pathology of Low-grade serous ovarian cancer.
Collapse
Affiliation(s)
| | - Hakimeh Zali
- Proteomics Research Center, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | | | - Babak Babaabasi
- Department of Genetics, Reproductive Biomedicine Research Center, Royan Institute, ACECR, Tehran, Iran
| |
Collapse
|
9
|
Shariat-Madar Z, Schmaier AH. Review: The plasma kallikrein/kinin and renin angiotensin systems in blood pressure regulation in sepsis. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519040100010101] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The hemodynamics of septic shock after endotoxinemiai s influenced by the plasma kallikrein/kinin and the renin angiotensin systems. In recent years, new information has improved understanding of the protein/biologically active peptide interactions between these two systems. The plasma kallikrein/kinin system, more commonly known as the contact system, has undergone a re-evaluation as to how it assembles on cell membranes for physiological and pathophysiological activation and as to its role in Gram-negative sepsis. It has been proposed that it counterbalances the plasma renin angiotensin system. Furthermore, more knowledge about the renin angiotensin system has become available on how it either opposes the actions of the kallikrein/kinin system or, in some cases, summates with it. Understanding the interactions between these two systems may lead to development of better pharmacological treatments for endotoxin-induced shock.
Collapse
Affiliation(s)
- Zia Shariat-Madar
- Departments of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA
| | - Alvin H. Schmaier
- Departments of Internal Medicine, The University of Michigan, Ann Arbor, Michigan, USA, , Department of Pathology, Hematology/Oncology Division, The University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
10
|
Wang NC, Matthews KA, Barinas-Mitchell EJ, Chang CCH, El Khoudary SR. Inflammatory/Hemostatic Biomarkers and Coronary Artery Calcium Progression in Women at Midlife (from the Study of Women's Health Across the Nation, Heart Study). Am J Cardiol 2016; 118:311-8. [PMID: 27289291 DOI: 10.1016/j.amjcard.2016.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 05/04/2016] [Accepted: 05/04/2016] [Indexed: 10/21/2022]
Abstract
It is unknown whether inflammatory/hemostatic biomarkers are associated with coronary artery calcium (CAC) progression. Our purpose was to evaluate the associations of baseline levels of C-reactive protein, fibrinogen, plasminogen activator inhibitor-1 (PAI-1), tissue plasminogen activator antigen, and circulating factor VII with CAC progression in healthy midlife women. Inflammatory/hemostatic biomarkers were measured at baseline. CAC was quantified by computed tomography scans at baseline and after 2.3 ± 0.5 years of follow-up. Significant CAC progression was defined as present if (1) follow-up CAC Agatston score was >0 if baseline CAC score = 0; (2) annualized change in CAC score was ≥10 if baseline CAC score >0 to <100; and (3) annualized percent change in CAC score was ≥10% if baseline CAC score ≥100. Extent of CAC progression was defined as [log(CAC(follow-up)+25) - log(CAC(baseline)+25)]/year. Logistic and linear regression models were used as appropriate, and the final models were adjusted for baseline CAC score, age, study site, race/ethnicity, menopausal status, sociodemographics, traditional cardiovascular disease (CVD) risk factors, family history of CVD, and CVD medication use. The study included 252 women (baseline age 51.2 ± 2.6 years; 67.5% white; 56.4% premenopausal or early perimenopausal). In final models, only log(PAI-1) was associated with presence of CAC progression (odds ratio 1.91, 95% CI 1.24 to 2.93; per 1 log unit increase in PAI-1; p = 0.003). In addition, higher log(PAI-1) was marginally associated with greater extent of CAC progression (p = 0.06). In conclusion, PAI-1 is associated with the presence of CAC progression in middle-aged women. Targeting PAI-1 may decrease atherogenesis beyond conventional CVD risk factors.
Collapse
|
11
|
Park BM, Cha SA, Lee SH, Kim SH. Angiotensin IV protects cardiac reperfusion injury by inhibiting apoptosis and inflammation via AT4R in rats. Peptides 2016; 79:66-74. [PMID: 27038740 DOI: 10.1016/j.peptides.2016.03.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 03/17/2016] [Accepted: 03/29/2016] [Indexed: 02/07/2023]
Abstract
Angiotensin IV (Ang IV) is formed by aminopeptidase N from Ang III by removing the first N-terminal amino acid. Previously, we reported that Ang III has some cardioprotective effects against global ischemia in Langendorff heart. However, it is not clear whether Ang IV has cardioprotective effects. The aim of the present study was to evaluate the effect of Ang IV on myocardial ischemia-reperfusion (I/R) injury in rats. Before ischemia, male Sprague-Dawley rats received Ang IV (1mg/kg/day) for 3 days. Anesthetized rats were subjected to 45min of ischemia by ligation of left anterior descending coronary artery followed by reperfusion and then, sacrificed 1 day or 1 week after reperfusion. Plasma creatine kinase (CK) and lactate dehydrogenase (LDH) concentrations, and infarct size were measured. Quantitative analysis of apoptotic and inflammatory proteins in ventricles were performed using Western blotting. Pretreatment with Ang IV attenuated I/R-induced increases in plasma CK and LDH levels, and infarct size, which were blunted by Ang IV receptor (AT4R) antagonist and but not by antagonist for AT1R, AT2R, or Mas receptor. I/R increased Bax, caspase-3 and caspase-9 protein levels, and decreased Bcl-2 protein level in ventricles, which were blunted by Ang IV. I/R-induced increases in TNF-α, MMP-9, and VCAM-1 protein levels in ventricles were also blunted by Ang IV. Ang IV increased the phosphorylation of Akt and mTOR. These effects were attenuated by co-treatment with AT4R antagonist or inhibitors of downstream signaling pathway. Myocardial dysfunction after reperfusion was improved by Ang IV. These results suggest that Ang IV has cardioprotective effect against I/R injury by inhibiting apoptosis via AT4R and PI3K-Akt-mTOR pathway.
Collapse
Affiliation(s)
- Byung Mun Park
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seung Ah Cha
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Sun Hwa Lee
- Internal Medicine, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Suhn Hee Kim
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea.
| |
Collapse
|
12
|
Meinert C, Gembardt F, Böhme I, Tetzner A, Wieland T, Greenberg B, Walther T. Identification of intracellular proteins and signaling pathways in human endothelial cells regulated by angiotensin-(1-7). J Proteomics 2015; 130:129-39. [PMID: 26388433 DOI: 10.1016/j.jprot.2015.09.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 08/21/2015] [Accepted: 09/12/2015] [Indexed: 10/23/2022]
Abstract
The study aimed to identify proteins regulated by the cardiovascular protective peptide angiotensin-(1-7) and to determine potential intracellular signaling cascades. Human endothelial cells were stimulated with Ang-(1-7) for 1 h, 3 h, 6 h, and 9 h. Peptide effects on intracellular signaling were assessed via antibody microarray, containing antibodies against 725 proteins. Bioinformatics software was used to identify affected intracellular signaling pathways. Microarray data was verified exemplarily by Western blot, Real-Time RT-PCR, and immunohistochemical studies. The microarray identified 110 regulated proteins after 1 h, 119 after 3 h, 31 after 6 h, and 86 after 9 h Ang-(1-7) stimulation. Regulated proteins were associated with high significance to several metabolic pathways like “Molecular Mechanism of Cancer” and “p53 signaling” in a time dependent manner. Exemplarily, Western blots for the E3-type small ubiquitin-like modifier ligase PIAS2 confirmed the microarray data and displayed a decrease by more than 50% after Ang-(1-7) stimulation at 1 h and 3 h without affecting its mRNA. Immunohistochemical studies with PIAS2 in human endothelial cells showed a decrease in cytoplasmic PIAS2 after Ang-(1-7) treatment. The Ang-(1-7) mediated decrease of PIAS2 was reproduced in other endothelial cell types. The results suggest that angiotensin-(1-7) plays a role in metabolic pathways related to cell death and cell survival in human endothelial cells.
Collapse
Affiliation(s)
- Christian Meinert
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, Universität Heidelberg, Germany; Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Florian Gembardt
- Division of Nephrology, Department of Internal Medicine III, Faculty of Medicine, Technische Universität Dresden, Germany
| | - Ilka Böhme
- Department of Obstetrics, Division of Women and Child Health, Universität Leipzig, Leipzig, Germany
| | - Anja Tetzner
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland; Department of Obstetrics, Division of Women and Child Health, Universität Leipzig, Leipzig, Germany
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, Universität Heidelberg, Germany
| | - Barry Greenberg
- Division of Cardiology, University of California, San Diego, USA
| | - Thomas Walther
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, Universität Heidelberg, Germany; Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland; Department of Obstetrics, Division of Women and Child Health, Universität Leipzig, Leipzig, Germany.
| |
Collapse
|
13
|
Senchenkova EY, Russell J, Esmon CT, Granger DN. Roles of Coagulation and fibrinolysis in angiotensin II-enhanced microvascular thrombosis. Microcirculation 2015; 21:401-7. [PMID: 24495184 DOI: 10.1111/micc.12120] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 01/29/2014] [Indexed: 11/28/2022]
Abstract
OBJECTIVE AngII-induced HTN is associated with accelerated thrombus development in arterioles. This study assessed the contributions of different components of the coagulation cascade and fibrinolysis to AngII-mediated microvascular thrombosis. METHODS Light/dye-induced thrombus formation (the time of onset and flow cessation) was quantified in cremaster muscle arterioles of AngII infused (two weeks) WT/AngII mice, EPCR-TgN, and mice deficient in PAI-1. WT/AngII mice were also treated with either tissue factor antibody, antithrombin III, heparin, hirudin, or murine APC. RESULTS TF immunoblockade or hirudin treatment did not prevent the AngII-induced acceleration of thrombosis. While antithrombin III treatment prevented the acceleration in both thrombus onset and flow cessation, heparin only improved the time for blood flow cessation. Neither WT mice treated with murine APC nor EPCR-TgN were protected against AngII-induced thrombus development. A similar lack of protection was noted in PAI-1deficient mice. CONCLUSION These findings implicate a role for thrombin generation pathway in the accelerated thrombosis induced by AngII and suggest that an impaired protein C pathway and increased PAI-1 do not make a significant contribution to this model of microvascular thrombosis.
Collapse
Affiliation(s)
- Elena Y Senchenkova
- Department of Molecular & Cellular Physiology, LSU Health Sciences Center, Shreveport, Louisiana, USA; Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, Saint-Petersburg, Russia
| | | | | | | |
Collapse
|
14
|
Park BM, Cha SA, Han BR, Kim SH. Angiotensin IV stimulates high atrial stretch-induced ANP secretion via insulin regulated aminopeptidase. Peptides 2015; 63:30-7. [PMID: 25451332 DOI: 10.1016/j.peptides.2014.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2014] [Revised: 10/27/2014] [Accepted: 10/27/2014] [Indexed: 01/11/2023]
Abstract
Angiotensin IV (Ang IV) is formed by aminopeptidase N (APN) from angiotensin III (Ang III) by removing the first N-terminal amino acid. Previously, we reported that angiotensin II (Ang II) inhibits atrial natriuretic peptide (ANP) secretion via angiotensin II type 1 receptor (AT1R). In contrast, angiotensin-(1-7) [Ang-(1-7)] and Ang III stimulate ANP secretion via Mas receptor (Mas R) and angiotensin II type 2 receptor (AT2R), respectively. However, it is not known whether there is any relationship between Ang IV and ANP secretion. Therefore, the aim of the present study was to determine the effect of Ang IV on ANP secretion and to find its downstream signaling pathway using in isolated perfused beating atria. Ang IV (0.1, 1 and 10μM) stimulated high atrial stretch-induced ANP secretion and ANP concentration in a dose-dependent manner. The augmented effect of Ang IV (1μM) on high atrial stretch-induced ANP secretion and concentration was attenuated by pretreatment with insulin-regulated aminopeptidase (IRAP) antagonist but not by AT1R or AT2R antagonist. Pretreatment with inhibitors of downstream signaling pathway including phosphatidylinositol 3-kinase (PI3K), protein kinase B (Akt) and mammalian target of rapamycin (mTOR) blocked Ang IV-induced ANP secretion and concentration. Therefore, these results suggest that Ang IV stimulates ANP secretion and concentration via IRAP and PI3K-Akt-mTOR pathway.
Collapse
Affiliation(s)
- Byung Mun Park
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Seung Ah Cha
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Bo Ram Han
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea
| | - Suhn Hee Kim
- Department of Physiology, Research Institute for Endocrine Sciences, Chonbuk National University Medical School, Jeonju, Republic of Korea.
| |
Collapse
|
15
|
Wright JW, Kawas LH, Harding JW. The development of small molecule angiotensin IV analogs to treat Alzheimer's and Parkinson's diseases. Prog Neurobiol 2014; 125:26-46. [PMID: 25455861 DOI: 10.1016/j.pneurobio.2014.11.004] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 11/17/2014] [Accepted: 11/19/2014] [Indexed: 02/07/2023]
Abstract
Alzheimer's (AD) and Parkinson's (PD) diseases are neurodegenerative diseases presently without effective drug treatments. AD is characterized by general cognitive impairment, difficulties with memory consolidation and retrieval, and with advanced stages episodes of agitation and anger. AD is increasing in frequency as life expectancy increases. Present FDA approved medications do little to slow disease progression and none address the underlying progressive loss of synaptic connections and neurons. New drug design approaches are needed beyond cholinesterase inhibitors and N-methyl-d-aspartate receptor antagonists. Patients with PD experience the symptomatic triad of bradykinesis, tremor-at-rest, and rigidity with the possibility of additional non-motor symptoms including sleep disturbances, depression, dementia, and autonomic nervous system failure. This review summarizes available information regarding the role of the brain renin-angiotensin system (RAS) in learning and memory and motor functions, with particular emphasis on research results suggesting a link between angiotensin IV (AngIV) interacting with the AT4 receptor subtype. Currently there is controversy over the identity of this AT4 receptor protein. Albiston and colleagues have offered convincing evidence that it is the insulin-regulated aminopeptidase (IRAP). Recently members of our laboratory have presented evidence that the brain AngIV/AT4 receptor system coincides with the brain hepatocyte growth factor/c-Met receptor system. In an effort to resolve this issue we have synthesized a number of small molecule AngIV-based compounds that are metabolically stable, penetrate the blood-brain barrier, and facilitate compromised memory and motor systems. These research efforts are described along with details concerning a recently synthesized molecule, Dihexa that shows promise in overcoming memory and motor dysfunctions by augmenting synaptic connectivity via the formation of new functional synapses.
Collapse
Affiliation(s)
- John W Wright
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA.
| | - Leen H Kawas
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| | - Joseph W Harding
- Departments of Psychology, Integrative Physiology and Neuroscience and Program in Biotechnology, Washington State University, Pullman, WA 99164-4820, USA; M3 Biotechnology, Inc., 4000 Mason Rd Suite 300, Box 352141, Seattle, WA 98195-2141, USA
| |
Collapse
|
16
|
Miller WM, Nori Janosz KE, Yanez J, McCullough PA. Effects of weight loss and pharmacotherapy on inflammatory markers of cardiovascular disease. Expert Rev Cardiovasc Ther 2014; 3:743-59. [PMID: 16076283 DOI: 10.1586/14779072.3.4.743] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Obesity is currently an epidemic, and the prevalence of cardiovascular risk factors is increasing dramatically as a result. Visceral adiposity is correlated with a proinflammatory and prothrombotic state that is believed to promote atherosclerosis and acute coronary syndromes. This article will review clinical trials on the effects of weight loss and pharmacotherapy on obesity associated inflammatory and thrombotic markers linked with cardiovascular disease.
Collapse
Affiliation(s)
- Wendy M Miller
- Beaumont Health Center, Weight Control Center, Division of Nutrition and Preventive Medicine, William Beaumont Hospital, Royal Oak, 4949 Coolidge Highway, Royal Oak, MI 48078, USA.
| | | | | | | |
Collapse
|
17
|
Wright JW, Kawas LH, Harding JW. A Role for the Brain RAS in Alzheimer's and Parkinson's Diseases. Front Endocrinol (Lausanne) 2013; 4:158. [PMID: 24298267 PMCID: PMC3829467 DOI: 10.3389/fendo.2013.00158] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Accepted: 10/09/2013] [Indexed: 12/30/2022] Open
Abstract
The brain renin-angiotensin system (RAS) has available the necessary functional components to produce the active ligands angiotensins II (AngII), angiotensin III, angiotensins (IV), angiotensin (1-7), and angiotensin (3-7). These ligands interact with several receptor proteins including AT1, AT2, AT4, and Mas distributed within the central and peripheral nervous systems as well as local RASs in several organs. This review first describes the enzymatic pathways in place to synthesize these ligands and the binding characteristics of these angiotensin receptor subtypes. We next discuss current hypotheses to explain the disorders of Alzheimer's disease (AD) and Parkinson's disease (PD), as well as research efforts focused on the use of angiotensin converting enzyme (ACE) inhibitors and angiotensin receptor blockers (ARBs), in their treatment. ACE inhibitors and ARBs are showing promise in the treatment of several neurodegenerative pathologies; however, there is a need for the development of analogs capable of penetrating the blood-brain barrier and acting as agonists or antagonists at these receptor sites. AngII and AngIV have been shown to play opposing roles regarding memory acquisition and consolidation in animal models. We discuss the development of efficacious AngIV analogs in the treatment of animal models of AD and PD. These AngIV analogs act via the AT4 receptor subtype which may coincide with the hepatocyte growth factor/c-Met receptor system. Finally, future research directions are described concerning new approaches to the treatment of these two neurological diseases.
Collapse
Affiliation(s)
- John W. Wright
- Departments of Psychology, Integrative Physiology and Neuroscience, Program in Biotechnology, Washington State University, Pullman, WA, USA
| | - Leen H. Kawas
- Departments of Psychology, Integrative Physiology and Neuroscience, Program in Biotechnology, Washington State University, Pullman, WA, USA
| | - Joseph W. Harding
- Departments of Psychology, Integrative Physiology and Neuroscience, Program in Biotechnology, Washington State University, Pullman, WA, USA
| |
Collapse
|
18
|
Feraco A, Armani A, Mammi C, Fabbri A, Rosano GMC, Caprio M. Role of mineralocorticoid receptor and renin-angiotensin-aldosterone system in adipocyte dysfunction and obesity. J Steroid Biochem Mol Biol 2013; 137:99-106. [PMID: 23454117 DOI: 10.1016/j.jsbmb.2013.02.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Revised: 02/07/2013] [Accepted: 02/20/2013] [Indexed: 12/13/2022]
Abstract
The mineralocorticoid receptor (MR) classically mediates aldosterone effects on salt homeostasis and blood pressure regulation in epithelial target tissues. In recent years, functional MRs have been identified in non classical targets of aldosterone actions, in particular in adipose tissue, where they mediate the effects of aldosterone and glucocorticoids in the control of adipogenesis, adipose expansion and its pro-inflammatory capacity. In this context, inappropriate MR activation has been demonstrated to be a causal factor in several pathologic conditions such as vascular inflammation, endothelial dysfunction, insulin resistance and obesity. The aim of this review is to summarize the latest developments in this rapidly developing field, and will focus on the role of MR and renin-angiotensin-aldosterone system (RAAS) as potential leading characters in the early steps of adipocyte dysfunction and obesity. Indeed modulation of MR activity in adipose tissue has promise as a novel therapeutic approach to treat obesity and its related metabolic complications. This article is part of a Special Issue entitled 'CSR 2013'.
Collapse
|
19
|
Hermanowicz JM, Hermanowicz A, Buczko P, Leszczynska A, Tankiewicz-Kwedlo A, Mogielnicki A, Buczko W. Aliskiren inhibits experimental venous thrombosis in two-kidney one- clip hypertensive rats. Thromb Res 2013; 131:e39-44. [DOI: 10.1016/j.thromres.2012.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2012] [Revised: 10/24/2012] [Accepted: 11/01/2012] [Indexed: 10/27/2022]
|
20
|
Brown NJ, Vaughan DE. The Renin-Angiotensin and fibrinolytic systems co-conspirators in the pathogenesis of ischemic cardiovascular disease. Trends Cardiovasc Med 2012; 6:239-43. [PMID: 21232303 DOI: 10.1016/s1050-1738(96)00091-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In vitro and in vivo data provide evidence for an interaction between the renin-angiotensin and fibrinolytic systems. Angiotensin-converting enzyme (ACE) is strategically poised to regulate this interaction. ACE catalyzes the conversion of angiotensin I to angiotensin II (Ang), and Ang II stimulates release of PAI-1, the major inhibitor of tissue-type plasminogen activator (t-PA) and urokinase in the vasculature. Conversely, ACE catalyzes the breakdown of bradykinin, a potent stimulus of t-PA secretion. This interaction between the renin-angiotensin and fibrinolytic systems may partially explain the clinical observation that stimulation or suppression of the renin-angiotensin system can alter the risk of ischemic cardiovascular events. © 1996, Elsevier Science Inc. (Trends Cardiovasc Med 1996;6:239-243).
Collapse
Affiliation(s)
- N J Brown
- Clinical Pharmacology Division, Departments of Medicine and Pharmacology, Vanderbilt University Medical Center,Nashville, TN 37232-1720,USA
| | | |
Collapse
|
21
|
Billings FT, Balaguer JM, C Y, Wright P, Petracek MR, Byrne JG, Brown NJ, Pretorius M. Comparative effects of angiotensin receptor blockade and ACE inhibition on the fibrinolytic and inflammatory responses to cardiopulmonary bypass. Clin Pharmacol Ther 2012; 91:1065-73. [PMID: 22549281 PMCID: PMC3822756 DOI: 10.1038/clpt.2011.356] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The effects of angiotensin-converting enzyme (ACE) inhibition and angiotensin II type 1 receptor blockade (ARB) on fibrinolysis and inflammation following cardiopulmonary bypass (CPB) are uncertain. This study tested the hypothesis that ACE inhibition enhances fibrinolysis and inflammation to greater extent than ARB in patients undergoing CPB.One week to five days prior to surgery, patients were randomized to ramipril 5mg/day,candesartan 16mg/day or placebo.ACE inhibition increased intraoperative bradykinin and tissue-type plasminogen activator (t-PA) concentrations compared to ARB. Both ACE inhibition and ARB decreased plasma transfusion compared to placebo, but only ACE inhibition decreased length of stay. Neither ACE inhibition nor ARB significantly affectedplasminogen activator inhibitor-1 (PAI-1), interleukin (IL)-6, IL-8, or IL-10 concentrations. ACE inhibition enhanced intraoperative fibrinolysis without increasing red cell transfusion risk. In contrast, neither ACE inhibition nor ARB affected the inflammatory response. ACE inhibitors and ARB may be safely continued until the day of surgery.
Collapse
Affiliation(s)
- F T Billings
- Department of Anesthesiology, Vanderbilt University Medical School, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Wright JW, Harding JW. The brain renin–angiotensin system: a diversity of functions and implications for CNS diseases. Pflugers Arch 2012; 465:133-51. [DOI: 10.1007/s00424-012-1102-2] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2012] [Revised: 03/20/2012] [Accepted: 03/30/2012] [Indexed: 12/14/2022]
|
23
|
Gembardt F, van Veghel R, Coffman TM, Schultheiss HP, Danser AHJ, Walther T. Hemodynamic effects of vasorelaxant compounds in mice lacking one, two or all three angiotensin II receptors. Hypertens Res 2012; 35:547-51. [DOI: 10.1038/hr.2012.5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
24
|
Wright JW, Harding JW. Brain renin-angiotensin—A new look at an old system. Prog Neurobiol 2011; 95:49-67. [DOI: 10.1016/j.pneurobio.2011.07.001] [Citation(s) in RCA: 147] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 06/27/2011] [Accepted: 07/03/2011] [Indexed: 12/15/2022]
|
25
|
Fogari R, Zoppi A, Mugellini A, Maffioli P, Lazzari P, Derosa G. Role of angiotensin II in plasma PAI-1 changes induced by imidapril or candesartan in hypertensive patients with metabolic syndrome. Hypertens Res 2011; 34:1321-6. [PMID: 21814211 DOI: 10.1038/hr.2011.137] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To evaluate the relationship between plasma plasminogen activator inhibitor-1 (PAI-1) and angiotensin II (Ang II) changes during treatment with imidapril and candesartan in hypertensive patients with metabolic syndrome. A total of 84 hypertensive patients with metabolic syndrome were randomized to imidapril 10 mg or candesartan 16 mg for 16 weeks. At weeks 4 and 8, there was a dose titration to imidapril 20 mg and candesartan 32 mg in nonresponders (systolic blood pressure (SBP) >140 and/or diastolic blood pressure (DBP) >90 mm Hg). We evaluated, at baseline and after 2, 4, 8, 12 and 16 weeks, clinic blood pressure, Ang II and PAI-1 antigen. Both imidapril and candesartan induced a similar SBP/DBP reduction (-19.4/16.8 and -19.5/16.3 mm Hg, respectively, P<0.001 vs. baseline). Both drugs decreased PAI-1 antigen after 4 weeks of treatment, but only the PAI-1 lowering effect of imidapril was sustained throughout the 16 weeks (-9.3 ng ml(-1), P<0.01 vs. baseline), whereas candesartan increased PAI-1 (+6.5 ng ml(-1), P<0.05 vs. baseline and P<0.01 vs. imidapril). Imidapril significantly decreased Ang II levels (-14.6 pg ml(-1) at week 16, P<0.05 vs. baseline), whereas candesartan increased them (+24.2 pg ml(-1), P<0.01 vs. baseline and vs. imidapril). In both groups there was a positive correlation between Ang II and PAI-1 changes (r=0.61, P<0.001 at week 16 for imidapril, and r=0.37, P<0.005 at week 16 for candesartan). Imidapril reduced plasma PAI-1 and Ang II levels, whereas candesartan increased them. This suggests that the different effect of angiotensin-converting enzyme inhibitors and Ang II blockers on Ang II production has a role in their different influence on fibrinolysis.
Collapse
Affiliation(s)
- Roberto Fogari
- Department of Internal Medicine and Therapeutics, Centro Ipertensione e Fisiopatologia Cardiovascolare, University of Pavia, Pavia, Italy.
| | | | | | | | | | | |
Collapse
|
26
|
Fogari R, Zoppi A, Salvadeo SAT, Mugellini A, Lazzari P, Santoro T, Derosa G. Fibrinolysis and insulin sensitivity in imidapril and candesartan (FISIC study) recipients with hypertension. Hypertens Res 2010; 34:509-15. [DOI: 10.1038/hr.2010.260] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
27
|
Naik P, Murumkar P, Giridhar R, Yadav MR. Angiotensin II receptor type 1 (AT1) selective nonpeptidic antagonists—A perspective. Bioorg Med Chem 2010; 18:8418-56. [DOI: 10.1016/j.bmc.2010.10.043] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2010] [Revised: 10/14/2010] [Accepted: 10/15/2010] [Indexed: 10/18/2022]
|
28
|
Senchenkova EY, Russell J, Almeida-Paula LD, Harding JW, Granger DN. Angiotensin II-mediated microvascular thrombosis. Hypertension 2010; 56:1089-95. [PMID: 20975035 DOI: 10.1161/hypertensionaha.110.158220] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Hypertension is associated with an increased risk of thrombosis that appears to involve an interaction between the renin-angiotensin system and hemostasis. In this study we determined whether angiotensin II-mediated thrombosis occurs in arterioles and/or venules and assessed the involvement of type 1 (AT₁), type 2 (AT₂), and type 4 (AT₄) angiotensin II receptors, as well as receptors for endothelin 1 and bradykinin 1 and 2 in angiotensin II-enhanced microvascular thrombosis. Thrombus development in mouse cremaster microvessels was quantified after light/dye injury using the time of onset of the thrombus and time to blood flow cessation. Wild-type and AT₁ receptor-deficient mice were implanted with an angiotensin II-loaded ALZET pump for 2 weeks. Angiotensin II administration in both wild-type and ATAT₁ receptor-deficient mice significantly accelerated thrombosis in arterioles. Genetic deficiency and pharmacological antagonism of AT₁ receptors did not alter the thrombosis response to angiotensin II. Isolated murine platelets aggregated in response to low (picomolar) but not high (nanomolar) concentrations of angiotensin II. The platelet aggregation response to angiotensin II depended on AT₁ receptors. Antagonism of AT₂ receptors in vivo significantly prolonged the onset of angiotensin II-enhanced thrombosis, whereas an AT₄ receptor antagonist prolonged the time to flow cessation. Selective antagonism of either endothelin 1 or bradykinin 1 receptors largely prevented both the onset and flow cessation responses to chronic angiotensin II infusion. Our findings indicate that angiotensin II induced hypertension is accompanied by enhanced thrombosis in arterioles, and this response is mediated by a mechanism that involves AT₂, AT₄, bradykinin 1, and endothelin 1 receptor-mediated signaling.
Collapse
Affiliation(s)
- Elena Y Senchenkova
- Department of Molecular and Cellular Physiology, Louisiana State University Health Science Center, 1501 Kings Hwy, Shreveport, LA 71130-3932.
| | | | | | | | | |
Collapse
|
29
|
Brown NJ. Review: Therapeutic potential of plasminogen activator inhibitor-1 inhibitors. Ther Adv Cardiovasc Dis 2010; 4:315-24. [DOI: 10.1177/1753944710379126] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the major physiological inhibitor of fibrinolysis and regulates cell migration and fibrosis. Preclinical studies using genetically altered mice and biological or small molecule inhibitors have elucidated a role for PAI-1 in the pathogenesis of thrombosis, vascular remodeling, renal injury, and initiation of diabetes. Inhibition of PAI-1 is a potential therapeutic strategy in these diseases.
Collapse
Affiliation(s)
- Nancy J. Brown
- 536 Robinson Research Building, Vanderbilt University Medical Center, Nashville, TN 37232-6602, USA,
| |
Collapse
|
30
|
Saad MAA, Abbas AM, Boshra V, Elkhateeb M, El Aal IA. Effect of angiotensin II type 1 receptor blocker, candesartan, and beta 1 adrenoceptor blocker, atenolol, on brain damage in ischemic stroke. ACTA ACUST UNITED AC 2010; 97:159-71. [PMID: 20511125 DOI: 10.1556/aphysiol.97.2010.2.2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
This work aims at studying the possible alteration of renal renin secretion after human ischemic stroke and correlating it to the post stroke neurological and renal function alterations using angiotensin II type 1(AT1) receptor blocker (ARB), candesartan, and beta 1 adrenoreceptor blocker atenolol, which inhibits renin secretion, in Wistar rats subjected to middle cerebral artery occlusion. Methods . This study comprised 21 patients with cerebral ischemic stroke. Seventeen normal persons were used for comparison. Recumbent and standing plasma renin activity (PRA), reflex plasma renin sensitivity, plasminogen activator inhibitor and creatinine clearance (Ccr) were estimated at admission and two weeks later. Moreover, 60 male Wistar rats were divided into two groups SHAM and ischemic. Each of the two groups was further subdivided into three subgroups, non-treated, atenolol treated, and candesartan treated. In all rats, mean arterial blood pressure (MAP), systolic blood pressure (SBP), diastolic blood pressure (DBP), pulse pressure (PP), heart rate (HR), neurobehavioral evaluation, Ccr, PRA, and infarct size were measured. Results . Together with the significant deterioration of the neurological score, focal cerebral ischemia in rats resulted in increased PRA and decreased glomerular filtration rate (GFR). In ischemic stroke patients, GFR was significantly decreased at admission and two weeks later, PRA increased at admission and two weeks later while plasma renin reflex secretion sensitivity had decreased significantly at admission relative to controls, but it increased significantly 2 weeks later. Atenolol caused significant improvement of the neurobehavioral score and renal function and decrease infarct size of rats subjected to focal cerebral ischemia whereas candesartan caused significant improvement of the neurobehavioral score and decreased infarct size with no significant change in GFR. Neither atenolol nor candesartan caused significant change in MAP, SBP, DBP, PP and HR Conclusion . (1) Ischemic stroke seems to be associated with a postischemic increase of the plasma renin secretion, which may increase the infarct size in the brain and may induce acute renal insufficiency. (2) This study confirms that Atenolol and ARBs could benefit ischemic stroke patients without altering blood pressure.
Collapse
Affiliation(s)
- M Ahdy A Saad
- Department of Pharmacology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | | | | | | | | |
Collapse
|
31
|
Affiliation(s)
- ANDREW S. GREENE
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
32
|
McKenzie TJ, Lillegard JB, Young WF, Thompson GB. Aldosteronomas--state of the art. Surg Clin North Am 2009; 89:1241-53. [PMID: 19836495 DOI: 10.1016/j.suc.2009.06.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Primary aldosteronism (PA) is the most common cause of secondary hypertension in nonsmokers. Widespread screening of unselected hypertensives has identified PA in as many as 15% of patients. With such screening efforts using the PAC/PRA ratio and PAC, the widespread prevalence of the disease has become apparent while the relative percentage of APA has decreased. PA is confirmed by demonstrating lack of aldosterone suppressibility with sodium loading. Subtype evaluation is best achieved with high resolution CT scanning and AVS in the appropriate setting. In patients with PA and a unilateral source of aldosterone excess, laparoscopic adrenalectomy is the treatment of choice with excellent outcomes and low morbidity as compared with older open approaches. Patients with IHA, or those not amenable or agreeable to surgery, are best managed with a MR antagonist.
Collapse
|
33
|
Yajima K, Shimada A, Hirose H, Oikawa Y, Yamada S, Meguro S, Irie J, Irie S. Effect on the atherogenic marker plasminogen activator inhibitor type-1 of addition of the ACE inhibitor imidapril to angiotensin II type 1 receptor antagonist therapy in hypertensive patients with abnormal glucose metabolism: a prospective cohort study in primary care. Clin Drug Investig 2009; 29:811-9. [PMID: 19888787 DOI: 10.2165/11530610-000000000-00000] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVE Renin-angiotensin system (RAS) inhibitors, such as angiotensin-converting enzyme (ACE) inhibitors and angiotensin II type 1 receptor antagonists (angiotensin receptor blockers [ARBs]), are recommended by the American Diabetes Association for blood pressure control and prevention or management of cardiovascular disease in patients with diabetes mellitus. However, some investigators have suggested that ARBs may increase the risk of myocardial infarction in hypertensive patients. Activation of the RAS is associated with an increased risk of ischaemic events. Angiotensin II stimulates the production of plasminogen activator inhibitor type-1 (PAI-1), a powerful predictor of cardiovascular disease. ACE inhibitors are reported to reduce PAI-1 levels and activity, while ARBs do not reduce or may even elevate levels of this atherogenic marker. The objective of this study was to determine whether the ACE inhibitor imidapril reduces PAI-1 levels in hypertensive patients already being treated with an ARB. METHODS This was a prospective cohort study carried out in primary care with a follow-up period of 6 months. Estimating the alpha error (p-value) at 0.05, the power of the test as 80%, and the difference in PAI-1 levels as 10 + or - 15 ng/mL, the required sample size was calculated to be 40. Participants were hypertensive patients taking ARBs for more than 8 weeks, and having dyslipidaemia, obesity or abnormal glucose metabolism. Imidapril 5-10 mg/day was prescribed for 6 months to reduce blood pressure to <130/80 mmHg. The main outcome measure, PAI-1 level, was measured before and 6 months after the addition of imidapril to ARBs in 21 subjects (13 men, eight women), all with abnormal glucose metabolism, nine with dyslipidaemia, and six who were obese. Bodyweight, body mass index, blood pressure, homeostasis model assessment of insulin resistance, glycosylated haemoglobin, creatinine, potassium, high sensitivity C-reactive protein (hs-CRP), and high molecular weight adiponectin levels were measured as secondary outcomes. RESULTS PAI-1 level was not significantly changed overall. Hs-CRP level was also not significantly changed; however, the high molecular weight adiponectin level was significantly increased (p = 0.044), especially in men (p = 0.026). There were no significant changes in the other outcomes measured. CONCLUSION The current study showed that imidapril added to ARBs did not decrease PAI-1 levels in hypertensive patients with abnormal glucose metabolism; however, this combination therapy significantly increased high molecular weight adiponectin levels in men.
Collapse
Affiliation(s)
- Ken Yajima
- Department of Internal Medicine, Federation of National Public Service Personnel Mutual Aid Associations, Tachikawa Hospital, 4-2-22 Nishikicho, Tachikawa,Tokyo 190-8531, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Pratte KA, Barón AE, Ogden LG, Hassell KL, Rewers M, Hokanson JE. Plasminogen activator inhibitor-1 is associated with coronary artery calcium in Type 1 diabetes. J Diabetes Complications 2009; 23:387-93. [PMID: 18768333 DOI: 10.1016/j.jdiacomp.2008.07.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2008] [Revised: 06/27/2008] [Accepted: 07/12/2008] [Indexed: 11/22/2022]
Abstract
BACKGROUND Elevated levels of plasminogen activator inhibitor-1 (PAI-1), the major inhibitor of fibrinolysis, is associated with coronary artery disease (CAD). This association may not be independent of factors related to insulin resistance (IR). Patients with Type 1 diabetes mellitus have increased CAD and an increase in sub-clinical CAD which develops earlier in life. It is not known if PAI-1 is associated with sub-clinical CAD in Type 1 diabetes or if this association is independent of IR. METHODS AND RESULTS Type 1 diabetes patients (n=560) and participants without diabetes (n=693) were assessed for coronary artery calcium (CAC), a surrogate for subclinical CAD, by electron-beam computed tomography. PAI-1 was associated with CAC in both Type 1 diabetes (OR=1.32, 95% CI=1.12-1.58) and non-diabetes (OR=1.34, 95% CI=1.13-1.58), after controlling for traditional risk factors not associated with IR. In Type 1 diabetes, the relationship between PAI-1 and CAC was strongest for younger participants (P=.02 for PAI-1-by-age interaction) after controlling for factors related to IR. PAI-1 was positively associated with CAC for Type 1 diabetes participants younger than 45 years of age. CONCLUSION PAI-1 levels are independently related to CAC in younger Type 1 diabetes participants. PAI-1 levels were not independently related to CAC in non-diabetes participants.
Collapse
Affiliation(s)
- Katherine A Pratte
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver, Denver, CO 80262, USA
| | | | | | | | | | | |
Collapse
|
35
|
Numaguchi Y, Ishii M, Kubota R, Morita Y, Yamamoto K, Matsushita T, Okumura K, Murohara T. Ablation of angiotensin IV receptor attenuates hypofibrinolysis via PAI-1 downregulation and reduces occlusive arterial thrombosis. Arterioscler Thromb Vasc Biol 2009; 29:2102-8. [PMID: 19745198 DOI: 10.1161/atvbaha.109.195057] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Reduced fibrinolytic activity is associated with adverse cardiovascular events. Although insulin-regulated aminopeptidase (IRAP) was recently identified as the angiotensin (Ang) IV receptor (AT4R), the impact of AngIV-AT4R signaling distal to AngII on the activation of type-1 plasminogen activator inhibitor (PAI-1) in the fibrinolytic process and subsequent formation of thrombosis remains unclarified. METHODS AND RESULTS To determine whether AngIV would inhibit fibrinolysis via PAI-1 activation and promote thrombosis, we evaluated the degree of fibrinolysis in thrombosis models and investigated the roles of AT4R after vascular injury using IRAP knockout mice (IRAP(-/-)). In endothelial cells from control mice (WT; C57Bl6/J), both AngII and AngIV treatments increased PAI-1 mRNA expression in a dose-dependent manner, whereas the response was blunted in endothelial cells from IRAP(-/-) mice. FeCl(3)-induced thrombosis was suppressed in the carotid arteries of IRAP(-/-) mice when compared with WT mice. Similarly, in a model of carotid artery ligation and cuff placement, IRAP(-/-) mice demonstrated accelerated fibrinolysis 7 days after surgery and reduced occlusive thrombosis with negative remodeling at 28 days. CONCLUSIONS AngIV-AT4R signaling has a key role in fibrinolysis and the subsequent formation of arterial thrombosis after vascular injury. AT4R may be a novel therapeutic target against cardiovascular disease.
Collapse
Affiliation(s)
- Yasushi Numaguchi
- Department of Medical Science of Proteases, Nagoya University School of Medicine, 65 Tsurumai, Showa-ku, Nagoya 466-8550, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Vanderheyden PML. From angiotensin IV binding site to AT4 receptor. Mol Cell Endocrinol 2009; 302:159-66. [PMID: 19071192 DOI: 10.1016/j.mce.2008.11.015] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2008] [Revised: 11/03/2008] [Accepted: 11/03/2008] [Indexed: 12/23/2022]
Abstract
One of the fragments of the cardiovascular hormone Angiotensin II incited the interest of several research groups. This 3-8 fragment, denoted as Angiotensin IV (Ang IV) causes a number of distinct biological effects (see Introduction), unlikely to be explained by its weak binding to AT(1) and/or AT(2) receptors. Moreover the discovery of high affinity [(125)I]-Ang IV binding sites and their particular tissue distribution led to the concept of the AT(4) receptor. An important breakthrough was achieved by defining the AT(4) receptor as the membrane-bound insulin-regulated aminopeptidase (IRAP). Crucial for the definition as a receptor the binding of the endogenous ligand(s) should be linked to particular cellular and/or biochemical processes. With this respect, cultured cells offer the possibility to study the presence of binding sites in conjunction with ligand induced signaling. This link is discussed for the AT(4) receptor by providing an overview of the cellular effects by AT(4) ligands.
Collapse
Affiliation(s)
- Patrick M L Vanderheyden
- Department of Molecular and Biochemical Pharmacology, Vrije Universiteit Brussel, Brussels, Belgium.
| |
Collapse
|
37
|
Soejima H, Ogawa H, Yasue H, Suefuji H, Kaikita K, Nishiyama K. Effects of imidapril therapy on endogenous fibrinolysis in patients with recent myocardial infarction. Clin Cardiol 2009; 20:441-5. [PMID: 9134274 PMCID: PMC6655695 DOI: 10.1002/clc.4960200507] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Treatment with an angiotensin-converting enzyme (ACE) inhibitor in patients with myocardial infarction has been shown to modify endogenous fibrinolysis. HYPOTHESIS We investigated the effects of the ACE inhibitor imidapril on endogenous fibrinolysis in association with the serum ACE activity. METHODS In a randomized, double-blind, placebo-controlled study beginning 4 weeks after uncomplicated myocardial infarction, 15 patients received imidapril (5 mg daily) (imidapril group) and another 15 received placebo therapy (placebo group) for 4 weeks. Blood sampling was performed before the start of administration and on Days 3, 7, and 28 after the start of administration. Serum ACE activity and plasma fibrinolytic variables [plasminogen activator inhibitor (PAI) activity, plasminogen activator inhibitor type 1 (PAI-1) antigen level, and tissue type plasminogen activator (TPA) antigen level] were measured. RESULTS There was no difference between the imidapril and placebo groups in serum ACE activity or plasma fibrinolytic variables before administration. Serum ACE activity decreased significantly on Days 3, 7, and 28 in the imidapril group. The decrease of PAI activity and PAI-1 antigen levels was significantly less on Days 7 and 28, but not on Day 3. The TPA antigen level in the imidapril group was unchanged. None of the parameters in the placebo group was changed. CONCLUSION The ACE inhibitor imidapril modified fibrinolysis, but the effects occurred after the inhibition of serum ACE activity.
Collapse
Affiliation(s)
- H Soejima
- Division of Cardiology, Kumamoto University School of Medicine, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Stanton A. Now that we have a direct renin inhibitor, what should we do with it? Curr Hypertens Rep 2008; 10:194-200. [DOI: 10.1007/s11906-008-0037-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
39
|
Renin-Angiotensin System. Cardiovasc Endocrinol 2008. [DOI: 10.1007/978-1-59745-141-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
|
40
|
Abstract
New components and functions of the renin-angiotensin system (RAS) are still being unravelled. The classical RAS as it looked in the middle 1970s consisted of circulating renin, acting on angiotensinogen to produce angiotensin I, which in turn was converted into angiotensin II (Ang II) by angiotensin-converting enzyme (ACE). Ang II, still considered the main effector of RAS was believed to act only as a circulating hormone via angiotensin receptors, AT1 and AT2. Since then, an expanded view of RAS has gradually emerged. Local tissue RAS systems have been identified in most organs. Recently, evidence for an intracellular RAS has been reported. The new expanded view of RAS therefore covers both endocrine, paracrine and intracrine functions. Other peptides of RAS have been shown to have biological actions; angiotensin 2-8 heptapeptide (Ang III) has actions similar to those of Ang II. Further, the angiotensin 3-8 hexapeptide (Ang IV) exerts its actions via insulin-regulated amino peptidase receptors. Finally, angiotensin 1-7 (Ang 1-7) acts via mas receptors. The discovery of another ACE2 was an important complement to this picture. The recent discovery of renin receptors has made our view of RAS unexpectedly complex and multilayered. The importance of RAS in cardiovascular disease has been demonstrated by the clinical benefits of ACE inhibitors and AT1 receptor blockers. Great expectations are now generated by the introduction of renin inhibitors. Indeed, RAS regulates much more and diverse physiological functions than previously believed.
Collapse
Affiliation(s)
- F Fyhrquist
- Minerva Institute for Medical Research and Department of Internal Medicine, Helsinki University Central Hospital, Helsinki, Finland.
| | | |
Collapse
|
41
|
Agirbasli M, Cincin A, Baykan OA. Short-term effects of angiotensin receptor blockers on blood pressure control, and plasma inflammatory and fibrinolytic parameters in patients taking angiotensin-converting enzyme inhibitors. J Renin Angiotensin Aldosterone Syst 2008; 9:22-6. [PMID: 18404606 DOI: 10.3317/jraas.2008.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
INTRODUCTION Angiotensin-converting enzyme (ACE) inhibitors reduce cardiovascular events in patients with established vascular disease and heart failure (HF). ACE-inhibitors have important effects on fibrinolytic balance, which may be the underlying mechanism for a reduction in cardiovascular events. Although angiotensin-receptor blockers (ARBs) offer greater tolerability than ACE-inhibitors, the major ARB trials have demonstrated a lack of reduction in myocardial infarction (MI) occurrence and mortality in contrast to ACE-inhibitors. In this study, we investigated the combined effects of ARBs and ACE-inhibitors on fibrinolytic and inflammatory parameters in patients with uncontrolled hypertension. METHODS Twenty-four patients with uncontrolled hypertension despite taking adequate doses of ACE-inhibitor therapy were selected. Patients were started on Candesartan 16 mg once a day. Plasma plasminogen activator inhibitor (PAI-1) antigen (Ag), tissue plasminogen activator (t-PA) Ag, thrombin-activatable fibrinolysis inhibitor (TAFI) % activity and high sensitivity C-reactive protein (hsCRP) levels, were measured during low salt intake at baseline and two weeks after therapy with an ARB. RESULTS Addition of ARB to the regimen reduced systolic (155+/-17 vs. 139+/-13, p<0.001), and diastolic (91+/-9 vs. 81+/-8, p<0.001) blood pressures (BP). No significant changes were observed in PAI-1 Ag (66+/-51 vs. 68+/-52, p=0.9), t-PA Ag (12.6+/-5.3 vs. 13.3+/-4.7, p=0.3), TAFI % activity (119+/-30 vs. 118+/-32, p=0.9) and hsCRP (3.9+/-3.4 vs. 3.6+/-3.6, p=0.7) levels after adding an ARB. CONCLUSIONS Combined ARB and ACE-inhibitor use provide better BP control without any detrimental effect in plasma inflammatory and fibrinolytic parameters.
Collapse
Affiliation(s)
- Mehmet Agirbasli
- Department of Cardiology, Marmara University School of Medicine, Istanbul, Turkey.
| | | | | |
Collapse
|
42
|
Ann Tallant E, Ferrario CM. Drug Evaluations Cardiovascular & Renal: Biology of angiotensin II receptor inhibition with a focus on losartan: A new drug for the treatment of hypertension. Expert Opin Investig Drugs 2008. [DOI: 10.1517/13543784.5.9.1201] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
43
|
Effects of aliskiren, a renin inhibitor, on biomarkers of platelet activity, coagulation and fibrinolysis in subjects with multiple risk factors for vascular disease. J Hum Hypertens 2008; 22:303-10. [DOI: 10.1038/jhh.2008.2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Wright JW, Yamamoto BJ, Harding JW. Angiotensin receptor subtype mediated physiologies and behaviors: new discoveries and clinical targets. Prog Neurobiol 2008; 84:157-81. [PMID: 18160199 PMCID: PMC2276843 DOI: 10.1016/j.pneurobio.2007.10.009] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2007] [Revised: 08/17/2007] [Accepted: 10/24/2007] [Indexed: 10/22/2022]
Abstract
The renin-angiotensin system (RAS) mediates several classic physiologies including body water and electrolyte homeostasis, blood pressure, cyclicity of reproductive hormones and sexual behaviors, and the regulation of pituitary gland hormones. These functions appear to be mediated by the angiotensin II (AngII)/AT(1) receptor subtype system. More recently, the angiotensin IV (AngIV)/AT(4) receptor subtype system has been implicated in cognitive processing, cerebroprotection, local blood flow, stress, anxiety and depression. There is accumulating evidence to suggest an inhibitory influence by AngII acting at the AT(1) subtype, and a facilitory role by AngIV acting at the AT(4) subtype, on neuronal firing rate, long-term potentiation, associative and spatial learning, and memory. This review initially describes the biochemical pathways that permit synthesis and degradation of active angiotensin peptides and three receptor subtypes (AT(1), AT(2) and AT(4)) thus far characterized. There is vigorous debate concerning the identity of the most recently discovered receptor subtype, AT(4). Descriptions of classic and novel physiologies and behaviors controlled by the RAS are presented. This review concludes with a consideration of the emerging therapeutic applications suggested by these newly discovered functions of the RAS.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, P.O. Box 644820, Pullman, WA 99164-4820, USA.
| | | | | |
Collapse
|
45
|
Boldt LH, Fraszl W, Röcker L, Schefold JC, Steinach M, Noack T, Gunga HC. Changes in the haemostatic system after thermoneutral and hyperthermic water immersion. Eur J Appl Physiol 2007; 102:547-54. [PMID: 18043935 DOI: 10.1007/s00421-007-0620-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2007] [Indexed: 11/29/2022]
Abstract
Warm water bathing is a popular recreational activity and is frequently used in rehabilitation medicine. Although well tolerated in most cases, there are reports indicating an increased risk of thrombotic events after hot tub bathing. The effects of a 45 min thermoneutral bath followed by a 50 min bath with increasing water temperature (maximum 41 degrees C) until reaching a body core temperature of 39 degrees C on factors of blood coagulation and fibrinolysis were studied in eight healthy male volunteers. Blood was obtained after a 45-min resting period as control and after the thermoneutral and hyperthermic bath as well as after another 45 min recovery period at the end of the study. Hyperthermic immersion (HI) lead to a shortening of activated partial thromboplastin time (aPTT) (P < 0.05). Fibrinogen concentration decreased immediately after HI (P < 0.05) but increased during recovery (P < 0.05). Plasminogen activator inhibitor (PAI) activity decreased during HI (P < 0.05), D-dimer concentration was not found to change. Thrombocyte count increased (P < 0.05) during HI. The increases in tissue-type plasminogen activator concentration as well as leucocyte count during HI were due to haemoconcentration. Prothrombin time, PAI-activity and granulocyte count decreased during thermoneutral immersion (P < 0.05). Warm water bathing leads to haemoconcentration and minimal activation of coagulation. The PAI-1 activity is decreased. A marked risk for thrombotic or bleeding complications during warm water bathing in healthy males could not be ascertained.
Collapse
Affiliation(s)
- Leif-Hendrik Boldt
- Department of Physiology, Center for Space Medicine Berlin, Charité, University Medicine Berlin, Campus Benjamin Franklin, Arnimallee 22, 14195, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
46
|
Involvement of insulin-regulated aminopeptidase in the effects of the renin–angiotensin fragment angiotensin IV: a review. Heart Fail Rev 2007; 13:321-37. [DOI: 10.1007/s10741-007-9062-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2007] [Accepted: 10/16/2007] [Indexed: 10/22/2022]
|
47
|
Vinh A, Widdop RE, Drummond GR, Gaspari TA. Chronic angiotensin IV treatment reverses endothelial dysfunction in ApoE-deficient mice. Cardiovasc Res 2007; 77:178-87. [DOI: 10.1093/cvr/cvm021] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
48
|
Brown NJ, Bradford J, Wang Z, Lea W, Ma L, Ma J, Vaughan DE, Fogo AB. Modulation of angiotensin II and norepinephrine-induced plasminogen activator inhibitor-1 expression by AT1a receptor deficiency. Kidney Int 2007; 72:72-81. [PMID: 17429342 DOI: 10.1038/sj.ki.5002268] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Angiotensin (Ang) II stimulates plasminogen activator inhibitor-1 (PAI-1) expression in many cell types by mechanisms that are cell-type specific. We measured effects of Ang II or norepinephrine on PAI-1 expression in wild type (WT) and Ang type-1a receptor knockout mice (AT(1a)-/-) in the presence or absence of the non-specific AT(1) antagonist losartan. Ang II and norepinephrine increased systolic blood pressure equally, whereas losartan decreased the pressor response of the former but not the latter in WT mice. In AT(1a)-/- mice, baseline systolic blood pressure was lower with no effect of Ang II, norepinephrine, or losartan. Ang II stimulated PAI-1 expression in the heart, aorta, and kidney and markedly in the liver of WT mice. In AT(1a)-/- mice, Ang II-stimulated PAI-1 was significantly attenuated compared with the WT in the heart and aorta but significantly enhanced in the kidney. Losartan decreased the induction in the aorta and liver of WT, and in the kidney and liver of AT(1a)-/- mice. Norepinephrine increased PAI-1 expression in WT heart and aorta, and in AT(1a)-/- heart, kidney, and liver with no effect of losartan. Renal PAI-1 expression correlated with AT(1b) receptor mRNA. We conclude that Ang II stimulates PAI-1 expression in part through the AT(1b) receptor in the kidney and liver. Further, norepinephrine induces PAI-1 expression in vivo with AT(1a) receptor deficiency modulating the effect.
Collapse
Affiliation(s)
- N J Brown
- Vanderbilt University Medical Center, Nashville, Tennessee 37232-6602, USA.
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Dimitropoulou C, Chatterjee A, McCloud L, Yetik-Anacak G, Catravas JD. Angiotensin, bradykinin and the endothelium. Handb Exp Pharmacol 2007:255-94. [PMID: 16999222 DOI: 10.1007/3-540-32967-6_8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Angiotensins and kinins are endogenous peptides with diverse biological actions; as such, they represent current and future targets of therapeutic intervention. The field of angiotensin biology has changed significantly over the last 50 years. Our original understanding of the crucial role of angiotensin II in the regulation of vascular tone and electrolyte homeostasis has been expanded to include the discovery of new angiotensins, their important role in cardiovascular inflammation and the development of clinically useful synthesis inhibitors and receptor antagonists. While less applied progress has been achieved in the kinin field, there are continuous discoveries in bradykinin physiology and in the complexity of kinin interactions with other proteins. The present review focuses on mechanisms and interactions of angiotensins and kinins that deal specifically with vascular endothelium.
Collapse
Affiliation(s)
- C Dimitropoulou
- Vascular Biology Center and Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta, GA 30912-2500, USA
| | | | | | | | | |
Collapse
|
50
|
Ruiz-Ortega M, Esteban V, Egido J. The regulation of the inflammatory response through nuclear factor-kappab pathway by angiotensin IV extends the role of the renin angiotensin system in cardiovascular diseases. Trends Cardiovasc Med 2007; 17:19-25. [PMID: 17210474 DOI: 10.1016/j.tcm.2006.10.003] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2006] [Accepted: 10/07/2006] [Indexed: 10/23/2022]
Abstract
The renin angiotensin system (RAS) participates in the pathogenesis of cardiovascular diseases. Although angiotensin II has been considered the effector peptide of RAS, accumulating evidence shows that other RAS peptides also posses important functions, some of them involved in cardiovascular pathology. Many studies support the importance of N-terminal angiotensin degradation product, angiotensin IV (AngIV), in the fields of cognition, renal metabolism, and pathophysiologic conditions. The novel data discussed here show that AngIV could contribute to cardiovascular damage. Angiotensin IV can be generated by degradation of angiotensin II, by aminopeptidase (AP) N, or by other proteases, which could be activated during tissue damage, suggesting that elevated AngIV levels can be found in pathologic conditions. Angiotensin IV binds to a specific receptor, AT(4), which has recently been identified as an insulin-regulated AP. In vascular cells, correspondence between AT(4) binding sites and insulin-regulated AP has been described. Angiotensin IV regulates cell growth in cardiac fibroblasts, endothelial cells, and vascular smooth muscle cells (VSMCs). In VSMC, AngIV, through AT(4), independently of AT(1) and AT(2) receptors, activates the nuclear factor-kappaB pathway and up-regulates several nuclear factor-kappaB-related genes, including the monocyte chemokine monocyte chemoattractant protein-1, the adhesion molecule intercellular adhesion molecule-1, and the cytokines interleukin 6 and tumor necrosis factor alpha. These data indicate that AngIV could be involved in the vascular inflammatory response. Thus, in endothelial cells and VSMC, AngIV up-regulates plasminogen activator inhibitor-1 expression and could participate in thrombus formation. These results reveal novel concepts of RAS in the cardiovascular system, suggesting that AngIV could play an active role in vascular diseases.
Collapse
Affiliation(s)
- Marta Ruiz-Ortega
- Cellular Biology in Renal Diseases Laboratory, Fundación Jiménez Diaz, Universidad Autónoma Madrid, Spain 28040.
| | | | | |
Collapse
|