1
|
Perry AS, Amancherla K, Huang X, Lance ML, Farber-Eger E, Gajjar P, Amrute J, Stolze L, Zhao S, Sheng Q, Joynes CM, Peng Z, Tanaka T, Drakos SG, Lavine KJ, Selzman C, Visker JR, Shankar TS, Ferrucci L, Das S, Wilcox J, Patel RB, Kalhan R, Shah SJ, Walker KA, Wells Q, Tucker N, Nayor M, Shah RV, Khan SS. Clinical-transcriptional prioritization of the circulating proteome in human heart failure. Cell Rep Med 2024; 5:101704. [PMID: 39226894 PMCID: PMC11524958 DOI: 10.1016/j.xcrm.2024.101704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 06/15/2024] [Accepted: 08/07/2024] [Indexed: 09/05/2024]
Abstract
Given expanding studies in epidemiology and disease-oriented human studies offering hundreds of associations between the human "ome" and disease, prioritizing molecules relevant to disease mechanisms among this growing breadth is important. Here, we link the circulating proteome to human heart failure (HF) propensity (via echocardiographic phenotyping and clinical outcomes) across the lifespan, demonstrating key pathways of fibrosis, inflammation, metabolism, and hypertrophy. We observe a broad array of genes encoding proteins linked to HF phenotypes and outcomes in clinical populations dynamically expressed at a transcriptional level in human myocardium during HF and cardiac recovery (several in a cell-specific fashion). Many identified targets do not have wide precedent in large-scale genomic discovery or human studies, highlighting the complementary roles for proteomic and tissue transcriptomic discovery to focus epidemiological targets to those relevant in human myocardium for further interrogation.
Collapse
Affiliation(s)
- Andrew S Perry
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Kaushik Amancherla
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Xiaoning Huang
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | | | - Eric Farber-Eger
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Priya Gajjar
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Junedh Amrute
- Cardiology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Lindsey Stolze
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Quanhu Sheng
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cassandra M Joynes
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Zhongsheng Peng
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Stavros G Drakos
- Division of Cardiovascular Medicine, University of Utah and Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), Salt Lake City, UT, USA
| | - Kory J Lavine
- Cardiology Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Craig Selzman
- Department of Cardiac Surgery, University of Utah School of Medicine, Division of Cardiothoracic Surgery, University of Utah and Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), Salt Lake City, UT, USA
| | - Joseph R Visker
- Division of Cardiovascular Medicine, University of Utah and Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), Salt Lake City, UT, USA
| | - Thirupura S Shankar
- Division of Cardiovascular Medicine, University of Utah and Nora Eccles Harrison Cardiovascular Research and Training Institute (CVRTI), Salt Lake City, UT, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Saumya Das
- Cardiovascular Division, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jane Wilcox
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ravi B Patel
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ravi Kalhan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Sanjiv J Shah
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Keenan A Walker
- Laboratory of Behavioral Neuroscience, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Quinn Wells
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | - Matthew Nayor
- Sections of Cardiovascular Medicine and Preventive Medicine and Epidemiology, Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Sadiya S Khan
- Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
2
|
Walker KE, Pasternak JA, Jones A, Mulligan MK, Van Goor A, Harding JCS, Lunney JK. Gene expression in heart, kidney, and liver identifies possible mechanisms underpinning fetal resistance and susceptibility to in utero PRRSV infection. Vet Microbiol 2024; 295:110154. [PMID: 38959808 DOI: 10.1016/j.vetmic.2024.110154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 06/12/2024] [Accepted: 06/15/2024] [Indexed: 07/05/2024]
Abstract
Porcine reproductive and respiratory syndrome (PRRS) is one of the costliest diseases to pork producers worldwide. We tested samples from the pregnant gilt model (PGM) to better understand the fetal response to in-utero PRRS virus (PRRSV) infection. Our goal was to identify critical tissues and genes associated with fetal resilience or susceptibility. Pregnant gilts (N=22) were infected with PRRSV on day 86 of gestation. At 21 days post maternal infection, the gilts and fetuses were euthanized, and fetal tissues collected. Fetuses were characterized for PRRS viral load in fetal serum and thymus, and preservation status (viable or meconium stained: VIA or MEC). Fetuses (N=10 per group) were compared: uninfected (UNIF; <1 log/µL PRRSV RNA), resilient (HV_VIA, >5 log virus/µL but viable), and susceptible (HV_MEC, >5 log virus/µL with MEC). Gene expression in fetal heart, kidney, and liver was investigated using NanoString transcriptomics. Gene categories investigated were hypothesized to be involved in fetal response to PRRSV infection: renin- angiotensin-aldosterone, inflammatory, transporter and metabolic systems. Following PRRSV infection, CCL5 increased expression in heart and kidney, and ACE2 decreased expression in kidney, each associated with fetal PRRS susceptibility. Liver revealed the most significant differential gene expression: CXCL10 decreased and IL10 increased indicative of immune suppression. Increased liver gene expression indicated potential associations with fetal PRRS susceptibility on several systems including blood pressure regulation (AGTR1), energy metabolism (SLC16A1 and SLC16A7), tissue specific responses (KL) and growth modulation (TGFB1). Overall, analyses of non-lymphoid tissues provided clues to mechanisms of fetal compromise following maternal PRRSV infection.
Collapse
Affiliation(s)
- K E Walker
- Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Services, Beltsville Agricultural Research Center, Beltsville, MD, United States; Department of Biology, Morgan State University, Baltimore, MD, United States
| | - J A Pasternak
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - A Jones
- Doctor of Veterinary Medicine program, St. George's University, True Blue, Grenada, West Indies
| | - M K Mulligan
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| | - A Van Goor
- United States Department of Agriculture, National Institute of Food and Agriculture, Columbia, MO, United States
| | - J C S Harding
- Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Dr., Saskatoon, Saskatchewan S7N 5B4, Canada
| | - J K Lunney
- Animal Parasitic Diseases Laboratory, United States Department of Agriculture, Agricultural Research Services, Beltsville Agricultural Research Center, Beltsville, MD, United States.
| |
Collapse
|
3
|
Kaschina E, Lauer D, Lange C, Unger T. Angiotensin AT 2 receptors reduce inflammation and fibrosis in cardiovascular remodeling. Biochem Pharmacol 2024; 222:116062. [PMID: 38369211 DOI: 10.1016/j.bcp.2024.116062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/04/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
The angiotensin AT2 receptor (AT2R), an important member of the "protective arm" of the renin-angiotensin system (RAS), has been recently defined as a therapeutic target in different pathological conditions. The AT2R activates complex signalling pathways linked to cellular proliferation, differentiation, anti-inflammation, antifibrosis, and induction or inhibition of apoptosis. The anti-inflammatory effect of AT2R activation is commonly associated with reduced fibrosis in different models. Current discoveries demonstrated a direct impact of AT2Rs on the regulation of cytokines, transforming growth factor beta1 (TGF-beta1), matrix metalloproteases (MMPs), and synthesis of the extracellular matrix components. This review article summarizes current knowledge on the AT2R in regard to immunity, inflammation and fibrosis in the heart and blood vessels. In particular, the differential influence of the AT2R on cardiovascular remodeling in preclinical models of myocardial infarction, heart failure and aneurysm formation are discussed. Overall, these studies demonstrate that AT2R stimulation represents a promising therapeutic approach to counteract myocardial and aortic damage in cardiovascular diseases.
Collapse
Affiliation(s)
- Elena Kaschina
- Charité - Universitätsmedizin Berlin, Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Berlin, Germany.
| | - Dilyara Lauer
- Charité - Universitätsmedizin Berlin, Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Berlin, Germany
| | - Christoph Lange
- Charité - Universitätsmedizin Berlin, Institute of Pharmacology, Max Rubner Center for Cardiovascular Metabolic Renal Research (MRC), Berlin, Germany
| | - Thomas Unger
- CARIM - School for Cardiovascular Diseases, Maastricht University, Maastricht, the Netherlands
| |
Collapse
|
4
|
Cohen-Segev R, Nativ O, Kinaneh S, Aronson D, Kabala A, Hamoud S, Karram T, Abassi Z. Effects of Angiotensin 1-7 and Mas Receptor Agonist on Renal System in a Rat Model of Heart Failure. Int J Mol Sci 2023; 24:11470. [PMID: 37511227 PMCID: PMC10380355 DOI: 10.3390/ijms241411470] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/09/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
Congestive heart failure (CHF) is often associated with impaired kidney function. Over- activation of the renin-angiotensin-aldosterone system (RAAS) contributes to avid salt/water retention and cardiac hypertrophy in CHF. While the deleterious effects of angiotensin II (Ang II) in CHF are well established, the biological actions of angiotensin 1-7 (Ang 1-7) are not fully characterized. In this study, we assessed the acute effects of Ang 1-7 (0.3, 3, 30 and 300 ng/kg/min, IV) on urinary flow (UF), urinary Na+ excretion (UNaV), glomerular filtration rate (GFR) and renal plasma flow )RPF) in rats with CHF induced by the placement of aortocaval fistula. Additionally, the chronic effects of Ang 1-7 (24 µg/kg/h, via intra-peritoneally implanted osmotic minipumps) on kidney function, cardiac hypertrophy and neurohormonal status were studied. Acute infusion of either Ang 1-7 or its agonist, AVE 0991, into sham controls, but not CHF rats, increased UF, UNaV, GFR, RPF and urinary cGMP. In the chronic protocols, untreated CHF rats displayed lower cumulative UF and UNaV than their sham controls. Chronic administration of Ang 1-7 and AVE 0991 exerted significant diuretic, natriuretic and kaliuretic effects in CHF rats, but not in sham controls. Serum creatinine and aldosterone levels were significantly higher in vehicle-treated CHF rats as compared with controls. Treatment with Ang 1-7 and AVE 0991 reduced these parameters to comparable levels observed in sham controls. Notably, chronic administration of Ang 1-7 to CHF rats reduced cardiac hypertrophy. In conclusion, Ang 1-7 exerts beneficial renal and cardiac effects in rats with CHF. Thus, we postulate that ACE2/Ang 1-7 axis represents a compensatory response to over-activity of ACE/AngII/AT1R system characterizing CHF and suggest that Ang 1-7 may be a potential therapeutic agent in this disease state.
Collapse
Affiliation(s)
- Ravit Cohen-Segev
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Omri Nativ
- Department of Urology, Rambam Health Center, Haifa 3109601, Israel
| | - Safa Kinaneh
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Doron Aronson
- Cardiology, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Aviva Kabala
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Shadi Hamoud
- Department of Internal Medicine E, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Tony Karram
- Vascular Surgery, Rambam Health Care Campus, Haifa 3109601, Israel
| | - Zaid Abassi
- Department of Physiology and Biophysics, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
- Laboratory Medicine, Rambam Health Care Campus, Haifa 31096, Israel
| |
Collapse
|
5
|
Oyama MA, Adin D. Toward quantification of loop diuretic responsiveness for congestive heart failure. J Vet Intern Med 2022; 37:12-21. [PMID: 36408832 PMCID: PMC9889629 DOI: 10.1111/jvim.16590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 11/11/2022] [Indexed: 11/22/2022] Open
Abstract
Diuretics, such as furosemide, are routinely administered to dogs with congestive heart failure (CHF). Traditionally, dose and determination of efficacy primarily are based on clinical signs rather than quantitative measures of drug action. Treatment of human CHF patients increasingly is guided by quantification of urine sodium concentration (uNa) and urine volume after diuretic administration. Use of these and other measures of diuretic responsiveness is associated with decreased duration of hospitalization, complication rates, future rehospitalization, and mortality. At their core, loop diuretics act through natriuresis, and attention to body sodium (Na) stores and handling offers insight into the pathophysiology of CHF and pharmacology of diuretics beyond what is achievable from clinical signs alone. Human patients with low diuretic responsiveness or diuretic resistance are at risk for difficult or incomplete decongestion that requires diuretic intensification or other remedial strategies. Identification of the specific etiology of resistance in a patient can help tailor personalized interventions. In this review, we advance the concept of loop diuretic responsiveness by highlighting Na and natriuresis. Specifically, we review body water homeostasis and congestion in light of the increasingly recognized role of interstitial Na, propose definitions for diuretic responsiveness and resistance in veterinary subjects, review relevant findings of recent studies, explain how the particular cause of resistance can guide treatment, and identify current knowledge gaps. We believe that a quantitative approach to loop diuretic usage primarily involving natriuresis will advance our understanding and care of dogs with CHF.
Collapse
Affiliation(s)
- Mark A. Oyama
- Clinical Sciences and Advanced MedicineUniversity of Pennsylvania, MJR‐VHUP‐CardiologyPhiladelphiaPennsylvaniaUSA
| | - Darcy Adin
- Large Animal Clinical SciencesUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
6
|
Abassi Z, Khoury EE, Karram T, Aronson D. Edema formation in congestive heart failure and the underlying mechanisms. Front Cardiovasc Med 2022; 9:933215. [PMID: 36237903 PMCID: PMC9553007 DOI: 10.3389/fcvm.2022.933215] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
Congestive heart failure (HF) is a complex disease state characterized by impaired ventricular function and insufficient peripheral blood supply. The resultant reduced blood flow characterizing HF promotes activation of neurohormonal systems which leads to fluid retention, often exhibited as pulmonary congestion, peripheral edema, dyspnea, and fatigue. Despite intensive research, the exact mechanisms underlying edema formation in HF are poorly characterized. However, the unique relationship between the heart and the kidneys plays a central role in this phenomenon. Specifically, the interplay between the heart and the kidneys in HF involves multiple interdependent mechanisms, including hemodynamic alterations resulting in insufficient peripheral and renal perfusion which can lead to renal tubule hypoxia. Furthermore, HF is characterized by activation of neurohormonal factors including renin-angiotensin-aldosterone system (RAAS), sympathetic nervous system (SNS), endothelin-1 (ET-1), and anti-diuretic hormone (ADH) due to reduced cardiac output (CO) and renal perfusion. Persistent activation of these systems results in deleterious effects on both the kidneys and the heart, including sodium and water retention, vasoconstriction, increased central venous pressure (CVP), which is associated with renal venous hypertension/congestion along with increased intra-abdominal pressure (IAP). The latter was shown to reduce renal blood flow (RBF), leading to a decline in the glomerular filtration rate (GFR). Besides the activation of the above-mentioned vasoconstrictor/anti-natriuretic neurohormonal systems, HF is associated with exceptionally elevated levels of atrial natriuretic peptide (ANP) and brain natriuretic peptide (BNP). However, the supremacy of the deleterious neurohormonal systems over the beneficial natriuretic peptides (NP) in HF is evident by persistent sodium and water retention and cardiac remodeling. Many mechanisms have been suggested to explain this phenomenon which seems to be multifactorial and play a major role in the development of renal hyporesponsiveness to NPs and cardiac remodeling. This review focuses on the mechanisms underlying the development of edema in HF with reduced ejection fraction and refers to the therapeutic maneuvers applied today to overcome abnormal salt/water balance characterizing HF.
Collapse
Affiliation(s)
- Zaid Abassi
- Department of Physiology, Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel
- Department of Laboratory Medicine, Rambam Health Care Campus, Haifa, Israel
- *Correspondence: Zaid Abassi,
| | - Emad E. Khoury
- Department of Physiology, Bruce Rappaport Faculty of Medicine, Technion–Israel Institute of Technology, Haifa, Israel
| | - Tony Karram
- Department of Vascular Surgery and Kidney Transplantation, Rambam Health Care Campus, Haifa, Israel
| | - Doron Aronson
- Department of Cardiology, Rambam Health Care Campus, Haifa, Israel
| |
Collapse
|
7
|
Shukla P, Chopada K, Sakure A, Hati S. Current Trends and Applications of Food-derived Antihypertensive
Peptides for the Management of Cardiovascular Disease. Protein Pept Lett 2022; 29:408-428. [DOI: 10.2174/0929866529666220106100225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 10/26/2021] [Accepted: 11/20/2021] [Indexed: 11/22/2022]
Abstract
Abstract:
Food derived Antihypertensive peptides is considered as a natural supplement for controlling the hypertension. Food protein not only serve as a macronutrient but also act as raw material for biosynthesis of physiologically active peptides. Food sources like milk and milk products, animal protein such as meat, chicken, fish, eggs and plant derived proteins from soy, rice, wheat, mushroom, pumpkins contain high amount of antihypertensive peptides. The food derived antihypertensive peptides has ability to supress the action of rennin and Angiotesin converting enzyme (ACE) which is mainly involved in regulation of blood pressure by RAS. The biosynthesis of endothelial nitric oxide synthase is also improved by ACE inhibitory peptides which increase the production of nitric oxide in vascular walls and encourage vasodilation. Interaction between the angiotensin II and its receptor is also inhibited by the peptides which help to reduce hypertension. This review will explore the novel sources and applications of food derived peptides for the management of hypertension.
Collapse
Affiliation(s)
- Pratik Shukla
- Department of Dairy Microbiology, SMC College of Dairy Science, Anand Agricultural University, Anand- 388110,
Gujarat, India
| | - Keval Chopada
- Department of Dairy Microbiology, SMC College of Dairy Science, Anand Agricultural University, Anand- 388110,
Gujarat, India
| | - Amar Sakure
- Department of Agricultural Biotechnology, Anand Agricultural University, Anand- 388110, Gujarat,
India
| | - Subrota Hati
- Department of Dairy Microbiology, SMC College of Dairy Science, Anand Agricultural University, Anand- 388110,
Gujarat, India
| |
Collapse
|
8
|
Effects of Renal Denervation on the Enhanced Renal Vascular Responsiveness to Angiotensin II in High-Output Heart Failure: Angiotensin II Receptor Binding Assessment and Functional Studies in Ren-2 Transgenic Hypertensive Rats. Biomedicines 2021; 9:biomedicines9121803. [PMID: 34944619 PMCID: PMC8698780 DOI: 10.3390/biomedicines9121803] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/16/2021] [Accepted: 11/27/2021] [Indexed: 02/01/2023] Open
Abstract
Detailed mechanism(s) of the beneficial effects of renal denervation (RDN) on the course of heart failure (HF) remain unclear. The study aimed to evaluate renal vascular responsiveness to angiotensin II (ANG II) and to characterize ANG II type 1 (AT1) and type 2 (AT2) receptors in the kidney of Ren-2 transgenic rats (TGR), a model of ANG II-dependent hypertension. HF was induced by volume overload using aorto-caval fistula (ACF). The studies were performed two weeks after RDN (three weeks after the creation of ACF), i.e., when non-denervated ACF TGR enter the decompensation phase of HF whereas those after RDN are still in the compensation phase. We found that ACF TGR showed lower renal blood flow (RBF) and its exaggerated response to intrarenal ANG II (8 ng); RDN further augmented this responsiveness. We found that all ANG II receptors in the kidney cortex were of the AT1 subtype. ANG II receptor binding characteristics in the renal cortex did not significantly differ between experimental groups, hence AT1 alterations are not responsible for renal vascular hyperresponsiveness to ANG II in ACF TGR, denervated or not. In conclusion, maintained renal AT1 receptor binding combined with elevated ANG II levels and renal vascular hyperresponsiveness to ANG II in ACF TGR influence renal hemodynamics and tubular reabsorption and lead to renal dysfunction in the high-output HF model. Since RDN did not attenuate the RBF decrease and enhanced renal vascular responsiveness to ANG II, the beneficial actions of RDN on HF-related mortality are probably not dominantly mediated by renal mechanism(s).
Collapse
|
9
|
Hashemi SMA, Thijssen M, Hosseini SY, Tabarraei A, Pourkarim MR, Sarvari J. Human gene polymorphisms and their possible impact on the clinical outcome of SARS-CoV-2 infection. Arch Virol 2021; 166:2089-2108. [PMID: 33934196 PMCID: PMC8088757 DOI: 10.1007/s00705-021-05070-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022]
Abstract
The SARS-CoV-2 pandemic has become one of the most serious health concerns globally. Although multiple vaccines have recently been approved for the prevention of coronavirus disease 2019 (COVID-19), an effective treatment is still lacking. Our knowledge of the pathogenicity of this virus is still incomplete. Studies have revealed that viral factors such as the viral load, duration of exposure to the virus, and viral mutations are important variables in COVID-19 outcome. Furthermore, host factors, including age, health condition, co-morbidities, and genetic background, might also be involved in clinical manifestations and infection outcome. This review focuses on the importance of variations in the host genetic background and pathogenesis of SARS-CoV-2. We will discuss the significance of polymorphisms in the ACE-2, TMPRSS2, vitamin D receptor, vitamin D binding protein, CD147, glucose-regulated protein 78 kDa, dipeptidyl peptidase-4 (DPP4), neuropilin-1, heme oxygenase, apolipoprotein L1, vitamin K epoxide reductase complex 1 (VKORC1), and immune system genes for the clinical outcome of COVID-19.
Collapse
Affiliation(s)
- Seyed Mohammad Ali Hashemi
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Microbiology, Golestan University of Medical Sciences, Gorgan, Iran
| | - Marijn Thijssen
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium
| | - Seyed Younes Hosseini
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Alijan Tabarraei
- Infectious Diseases Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Mahmoud Reza Pourkarim
- Laboratory for Clinical and Epidemiological Virology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, 3000 Leuven, Belgium
- Health Policy Research Centre, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jamal Sarvari
- Department of Bacteriology and Virology, Shiraz University of Medical Sciences, Shiraz, Iran
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
Paniri A, Hosseini MM, Akhavan-Niaki H. First comprehensive computational analysis of functional consequences of TMPRSS2 SNPs in susceptibility to SARS-CoV-2 among different populations. J Biomol Struct Dyn 2021; 39:3576-3593. [PMID: 32410502 PMCID: PMC7284145 DOI: 10.1080/07391102.2020.1767690] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 05/04/2020] [Indexed: 12/23/2022]
Abstract
Current SARS-CoV-2 pandemy mortality created the hypothesis that some populations may be more susceptible to SARS-CoV-2. TMPRSS2 encodes a transmembrane serine protease which plays a crucial role in SARS-CoV-2 cell entry. Single nucleotide polymorphisms (SNPs) in TMPRSS2 might influence SARS-CoV2 entry into the cell. This study aimed to investigate the impact of SNPs on TMPRSS2 function and structure. In silico tools such as Ensembl, Gtex, ExPASY 2, GEPIA, CCLE, KEGG and GO were engaged to characterize TMPRSS2 and its expression profile. The functional effects of SNPs were analyzed by PolyPhen-2, PROVEN, SNAP2, SIFT and HSF. Also, Phyre2, GOR IV and PSIPRED were used to predict the secondary structure of TMPRSS2. Moreover, post-translational modification (PTM) and secretory properties were analyzed through Modpredand Phobius, respectively. Finally, miRNA profiles were investigated by PolymiRTS and miRSNPs. Out of 11,184 retrieved SNPs from dbSNP, 92 showed a different frequency between Asians and other populations. Only 21 SNPs affected the function and structure of TMPRSS2 by influencing the protein folding, PTM, splicing and miRNA function. Particularly, rs12329760 may create a de novo pocket protein. rs875393 can create a donor site, silencer and broken enhancer motifs. rs12627374 affects a wide spectrum of miRNAs profile. This study highlighted the role of TMPRSS2 SNPs and epigenetic mechanisms especially non-coding RNAs in appearance of different susceptibility to SARS-CoV-2 among different populations. Also, this study could pave the way to potential therapeutic implication of TMPRSS2 in designing antiviral drugs.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Alireza Paniri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Genetics Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Haleh Akhavan-Niaki
- Genetics Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
- Zoonoses Research Center, Pasteur Institute of Iran, Amol, Iran
| |
Collapse
|
11
|
Khoury EE, Knaney Y, Fokra A, Kinaneh S, Azzam Z, Heyman SN, Abassi Z. Pulmonary, cardiac and renal distribution of ACE2, furin, TMPRSS2 and ADAM17 in rats with heart failure: Potential implication for COVID-19 disease. J Cell Mol Med 2021; 25:3840-3855. [PMID: 33660945 PMCID: PMC8014258 DOI: 10.1111/jcmm.16310] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 12/20/2022] Open
Abstract
Congestive heart failure (CHF) is often associated with kidney and pulmonary dysfunction. Activation of the renin-angiotensin-aldosterone system (RAAS) contributes to avid sodium retention, cardiac hypertrophy and oedema formation, including lung congestion. While the status of the classic components of RAAS such as renin, angiotensin converting enzyme (ACE), angiotensin II (Ang II) and angiotensin II receptor AT-1 is well studied in CHF, the expression of angiotensin converting enzyme-2 (ACE2), a key enzyme of angiotensin 1-7 (Ang 1-7) generation in the pulmonary, cardiac and renal systems has not been studied thoroughly in this clinical setting. This issue is of a special interest as Ang 1-7 counterbalance the vasoconstrictory, pro-inflammatory and pro-proliferative actions of Ang II. Furthermore, CHF predisposes to COVID-19 disease severity, while ACE2 also serves as the binding domain of SARS-CoV-2 in human host-cells, and acts in concert with furin, an important enzyme in the synthesis of BNP in CHF, in permeating viral functionality along TMPRSST2. ADAM17 governs ACE2 shedding from cell membranes. Therefore, the present study was designed to investigate the expression of ACE2, furin, TMPRSS2 and ADAM17 in the lung, heart and kidneys of rats with CHF to understand the exaggerated susceptibility of clinical CHF to COVID-19 disease. Heart failure was induced in male Sprague Dawley rats by the creation of a surgical aorto-caval fistula. Sham-operated rats served as controls. One week after surgery, the animals were subdivided into compensated and decompensated CHF according to urinary sodium excretion. Both groups and their controls were sacrificed, and their hearts, lungs and kidneys were harvested for assessment of tissue remodelling and ACE2, furin, TMPRSS2 and ADAM17 immunoreactivity, expression and immunohistochemical staining. ACE2 immunoreactivity and mRNA levels increased in pulmonary, cardiac and renal tissues of compensated, but not in decompensated CHF. Furin immunoreactivity was increased in both compensated and decompensated CHF in the pulmonary, cardiac tissues and renal cortex but not in the medulla. Interestingly, both the expression and abundance of pulmonary, cardiac and renal TMPRSS2 decreased in CHF in correlation with the severity of the disease. Pulmonary, cardiac and renal ADAM17 mRNA levels were also downregulated in decompensated CHF. Circulating furin levels increased in proportion to CHF severity, whereas plasma ACE2 remained unchanged. In summary, ACE2 and furin are overexpressed in the pulmonary, cardiac and renal tissues of compensated and to a lesser extent of decompensated CHF as compared with their sham controls. The increased expression of the ACE2 in heart failure may serve as a compensatory mechanism, counterbalancing the over-activity of the deleterious isoform, ACE. Downregulated ADAM17 might enhance membranal ACE2 in COVID-19 disease, whereas the suppression of TMPRSS2 in CHF argues against its involvement in the exaggerated susceptibility of CHF patients to SARS-CoV2.
Collapse
Affiliation(s)
- Emad E. Khoury
- Department of Physiology and BiophysicsRappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Yara Knaney
- Department of Physiology and BiophysicsRappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Ahmad Fokra
- Department of Physiology and BiophysicsRappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Safa Kinaneh
- Department of Physiology and BiophysicsRappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
| | - Zaher Azzam
- Department of Internal MedicineRambam Medical CenterHaifaIsrael
| | - Samuel N. Heyman
- Department of MedicineHadassah Hebrew University HospitalJerusalemIsrael
| | - Zaid Abassi
- Department of Physiology and BiophysicsRappaport Faculty of MedicineTechnion‐Israel Institute of TechnologyHaifaIsrael
- Laboratory MedicineRambam Medical CenterHaifaIsrael
| |
Collapse
|
12
|
Amirfakhryan H, Safari F. Outbreak of SARS-CoV2: Pathogenesis of infection and cardiovascular involvement. Hellenic J Cardiol 2021; 62:13-23. [PMID: 32522617 PMCID: PMC7275139 DOI: 10.1016/j.hjc.2020.05.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/22/2020] [Accepted: 05/28/2020] [Indexed: 02/06/2023] Open
Abstract
Since the new severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) has emerged from China, the infection (novel corona virus disease-2019, COVID-19) has affected many countries and led to many deaths worldwide. Like SARS-CoV, angiotencin converting enzyme (ACE)2 as a functional receptor for SARS-CoV2 is essential for the virus to make an entry into the cell. ACE2 is a part of Renin-Angiotensin-Aldosterone System, which is expressed in several organs that opposes the angiotensin (Ang) II functions by converting Ang II to Ang (1-7), the one with vasodilation effects. The death rate of COVID-19 is estimated to be approximately 3.4%; however, some comorbid conditions like underlying cardiovascular disease, hypertension, and diabetes increase the risk of mortality. In addition, cardiovascular involvement as a complication of SARS-CoV2 could be direct through either ACE2 receptors that are expressed tremendously in the heart, or by the surge of different cytokines or by acute respiratory distress syndrome-induced hypoxia. Traditional risk factors could aggravate the process of COVID-19 infection that urges the triage of these high-risk patients for SARS-CoV2. Currently, there is no effective, proven treatment or vaccination for COVID-19, but many investigators are struggling to find a treatment strategy as soon as possible. Some potential medications like chloroquine by itself or in combination with azithromycin and some protease inhibitors used for the treatment of COVID-19 have cardiovascular adverse effects, which should be kept in mind while the patients taking these medications are being closely monitored.
Collapse
Affiliation(s)
- Hamideh Amirfakhryan
- University of South Wales, Faculty of Health Science, Preventative Cardiovascular Medicine, UK.
| | - Fatemeh Safari
- University of Alberta, Edmonton, Faculty of Medicine, AB, Canada
| |
Collapse
|
13
|
Paniri A, Akhavan-Niaki H. Emerging role of IL-6 and NLRP3 inflammasome as potential therapeutic targets to combat COVID-19: Role of lncRNAs in cytokine storm modulation. Life Sci 2020; 257:118114. [PMID: 32693241 PMCID: PMC7368418 DOI: 10.1016/j.lfs.2020.118114] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/13/2020] [Indexed: 02/07/2023]
Abstract
The world has witnessed a high morbidity and mortality caused by SARS-CoV-2, and global death toll is still rising. Exaggerated inflammatory responses are thought to be more responsible for infiltrated immune cells accumulation, organ damage especially lung, dyspnea, and respiratory failure rather than direct effect of viral replication. IL-6 and NLRP3 inflammasome are the major immune components in immune responses stimulation upon pathogen infection. It's noteworthy that the function and expression of these components are remarkably influenced by non-coding RNAs including long non-coding RNAs. Given the potential role of these components in organ damage and pathological manifestations of patients infected with COVID-19, their blockage might be a hopeful and promising treatment strategy. Notably, more study on long non-coding RNAs involved in inflammatory responses could elevate the efficacy of anti-inflammatory therapy. In this review we discuss the potential impact of IL-6 and NLRP3 inflammasome blocker drugs on inflammatory responses, viral clearance, and pathological and clinical manifestations. Collectively, anti-inflammatory strategy might pave the way to diminish clinical and pathological manifestations and thereby discharging patients infected with COVID-19 from hospital.
Collapse
Affiliation(s)
- Alireza Paniri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran; Genetics Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Haleh Akhavan-Niaki
- Genetics Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran; Zoonoses Research Center, Pasteur Institute of Iran, Amol, Iran.
| |
Collapse
|
14
|
Kimura H, Francisco D, Conway M, Martinez FD, Vercelli D, Polverino F, Billheimer D, Kraft M. Type 2 inflammation modulates ACE2 and TMPRSS2 in airway epithelial cells. J Allergy Clin Immunol 2020; 146:80-88.e8. [PMID: 32422146 PMCID: PMC7227558 DOI: 10.1016/j.jaci.2020.05.004] [Citation(s) in RCA: 252] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 05/05/2020] [Accepted: 05/07/2020] [Indexed: 01/04/2023]
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has dramatically changed our world, country, communities, and families. There is controversy regarding risk factors for severe COVID-19 disease. It has been suggested that asthma and allergy are not highly represented as comorbid conditions associated with COVID-19. Objective Our aim was to extend our work in IL-13 biology to determine whether airway epithelial cell expression of 2 key mediators critical for SARS-CoV-2 infection, namely, angiotensin-converting enzyme 2 (ACE2) and transmembrane protease, serine 2 (TMPRSS2), are modulated by IL-13. Methods We determined effects of IL-13 treatment on ACE2 and TMPRSS2 expression ex vivo in primary airway epithelial cells from participants with and without type 2 asthma obtained by bronchoscopy. We also examined expression of ACE2 and TMPRSS2 in 2 data sets containing gene expression data from nasal and airway epithelial cells from children and adults with asthma and allergic rhinitis. Results IL-13 significantly reduced ACE2 and increased TMPRSS2 expression ex vivo in airway epithelial cells. In 2 independent data sets, ACE2 expression was significantly reduced and TMPRSS2 expression was significantly increased in the nasal and airway epithelial cells in type 2 asthma and allergic rhinitis. ACE2 expression was significantly negatively associated with type 2 cytokines, whereas TMPRSS2 expression was significantly positively associated with type 2 cytokines. Conclusion IL-13 modulates ACE2 and TMPRSS2 expression in airway epithelial cells in asthma and atopy. This deserves further study with regard to any effects that asthma and atopy may render in the setting of COVID-19 infection.
Collapse
Affiliation(s)
- Hiroki Kimura
- Department of Medicine, College of Medicine Tucson, University of Arizona Health Sciences, Tucson, Ariz
| | - Dave Francisco
- Department of Medicine, College of Medicine Tucson, University of Arizona Health Sciences, Tucson, Ariz
| | - Michelle Conway
- Department of Medicine, College of Medicine Tucson, University of Arizona Health Sciences, Tucson, Ariz
| | - Fernando D Martinez
- Asthma and Airway Disease Research Center, College of Medicine Tucson, University of Arizona Health Sciences, Tucson, Ariz
| | - Donata Vercelli
- Asthma and Airway Disease Research Center, College of Medicine Tucson, University of Arizona Health Sciences, Tucson, Ariz
| | - Francesca Polverino
- Department of Medicine, College of Medicine Tucson, University of Arizona Health Sciences, Tucson, Ariz
| | - Dean Billheimer
- Department of Epidemiology and Biostatistics, Mel and Enid Zuckerman College of Public Health, University of Arizona, Tucson, Ariz
| | - Monica Kraft
- Department of Medicine, College of Medicine Tucson, University of Arizona Health Sciences, Tucson, Ariz.
| |
Collapse
|
15
|
Silva FH, Veiga FJR, Mora AG, Heck RS, De Oliveira CC, Gambero A, Franco-Penteado CF, Antunes E, Gardner JD, Priviero FBM, Claudino MA. A novel experimental model of erectile dysfunction in rats with heart failure using volume overload. PLoS One 2017; 12:e0187083. [PMID: 29095897 PMCID: PMC5667775 DOI: 10.1371/journal.pone.0187083] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 10/15/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Patients with heart failure (HF) display erectile dysfunction (ED). However, the pathophysiology of ED during HF remains poorly investigated. OBJECTIVE This study aimed to characterize the aortocaval fistula (ACF) rat model associated with HF as a novel experimental model of ED. We have undertaken molecular and functional studies to evaluate the alterations of the nitric oxide (NO) pathway, autonomic nervous system and oxidative stress in the penis. METHODS Male rats were submitted to ACF for HF induction. Intracavernosal pressure in anesthetized rats was evaluated. Concentration-response curves to contractile (phenylephrine) and relaxant agents (sodium nitroprusside; SNP), as well as to electrical field stimulation (EFS), were obtained in the cavernosal smooth muscle (CSM) strips from sham and HF rats. Protein expression of endothelial NO synthase (eNOS) and neuronal NO synthase (nNOS) and phosphodiestarese-5 in CSM were evaluated, as well as NOX2 (gp91phox) and superoxide dismutase (SOD) mRNA expression. SOD activity and thiobarbituric acid reactive substances (TBARs) were also performed in plasma. RESULTS HF rats display erectile dysfunction represented by decreased ICP responses compared to sham rats. The neurogenic contractile responses elicited by EFS were greater in CSM from the HF group. Likewise, phenylephrine-induced contractions were greater in CSM from HF rats. Nitrergic response induced by EFS were decreased in the cavernosal tissue, along with lower eNOS, nNOS and phosphodiestarese-5 protein expressions. An increase of NOX2 and SOD mRNA expression in CSM and plasma TBARs of HF group were detected. Plasma SOD activity was decreased in HF rats. CONCLUSION ED in HF rats is associated with decreased NO bioavailability in erectile tissue due to eNOS/nNOS dowregulation and NOX2 upregulation, as well as hypercontractility of the penis. This rat model of ACF could be a useful tool to evaluate the molecular alterations of ED associated with HF.
Collapse
Affiliation(s)
- Fábio Henrique Silva
- Hematology and Hemotherapy Center, University of Campinas, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Frederico José Reis Veiga
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| | - Aline Gonçalves Mora
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| | - Rodrigo Sader Heck
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| | - Caroline Candida De Oliveira
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| | - Alessandra Gambero
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| | - Carla Fernanda Franco-Penteado
- Hematology and Hemotherapy Center, University of Campinas, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Edson Antunes
- Department of Pharmacology, Faculty of Medical Sciences, University of Campinas, Campinas, SP, Brazil
| | - Jason D. Gardner
- Department of Physiology, Louisiana State University Health Sciences Center, New Orleans, LA, United States of America
| | | | - Mário Angelo Claudino
- Laboratory of Multidisciplinary Research, São Francisco University Medical School, Bragança Paulista, SP, Brazil
| |
Collapse
|
16
|
Azzam ZS, Kinaneh S, Bahouth F, Ismael-Badarneh R, Khoury E, Abassi Z. Involvement of Cytokines in the Pathogenesis of Salt and Water Imbalance in Congestive Heart Failure. Front Immunol 2017; 8:716. [PMID: 28674538 PMCID: PMC5474564 DOI: 10.3389/fimmu.2017.00716] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 06/02/2017] [Indexed: 12/28/2022] Open
Abstract
Congestive heart failure (CHF) has become a major medical problem in the western world with high morbidity and mortality rates. CHF adversely affects several systems, mainly the kidneys and the lungs. While the involvement of the renin-angiotensin-aldosterone system and the sympathetic nervous system in the progression of cardiovascular, pulmonary, and renal dysfunction in experimental and clinical CHF is well established, the importance of pro-inflammatory mediators in the pathogenesis of this clinical setting is still evolving. In this context, CHF is associated with overexpression of pro-inflammatory cytokines, such as tumor necrosis factor-α, interleukin (IL)-1, and IL-6, which are activated in response to environmental injury. This family of cytokines has been implicated in the deterioration of CHF, where it plays an important role in initiating and integrating homeostatic responses both at the myocardium and circulatory levels. We and others showed that angiotensin II decreased the ability of the lungs to clear edema and enhanced the fibrosis process via phosphorylation of the mitogen-activated protein kinases p38 and p42/44, which are generally involved in cellular responses to pro-inflammatory cytokines. Literature data also indicate the involvement of these effectors in modulating ion channel activity. It has been reported that in heart failure due to mitral stenosis; there were varying degrees of vascular and other associated parenchymal changes such as edema and fibrosis. In this review, we will discuss the effects of cytokines and other inflammatory mediators on the kidneys and the lungs in heart failure; especially their role in renal and alveolar ion channels activity and fluid balance.
Collapse
Affiliation(s)
- Zaher S. Azzam
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
- Internal Medicine “B”, Rambam Health Care Campus, Haifa, Israel
| | - Safa Kinaneh
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Fadel Bahouth
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Reem Ismael-Badarneh
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Emad Khoury
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| | - Zaid Abassi
- Department of Physiology and Biophysics, Technion, Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
17
|
Angiotensin-converting enzyme 2 inhibits high-mobility group box 1 and attenuates cardiac dysfunction post-myocardial ischemia. J Mol Med (Berl) 2016; 94:37-49. [PMID: 26498282 DOI: 10.1007/s00109-015-1356-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 08/03/2015] [Accepted: 09/24/2015] [Indexed: 12/23/2022]
Abstract
High-mobility group box 1 (HMGB1) triggers and amplifies inflammation cascade following ischemic injury, and its elevated levels are associated with adverse clinical outcomes in patients with myocardial infarction (MI). Angiotensin-converting enzyme 2 (ACE2), a key member of vasoprotective axis of the renin-angiotensin system (RAS), regulates cardiovascular functions and exerts beneficial effects in cardiovascular disease. However, the association between HMGB1 and ACE2 has not been studied. We hypothesized that overexpression of ACE2 provides cardioprotective effects against MI via inhibiting HMGB1 and inflammation. ACE2 knock-in (KI) mice and littermate wild-type (WT) controls were subjected to either sham or coronary artery ligation surgery to induce MI. Heart function was assessed 4 weeks after surgery using echocardiography and Millar catheterization. Tissues were collected for histology and analysis of the expression of HMGB1, RAS components, and inflammatory cytokines. ACE2 in the heart of the ACE2 KI mice was 58-fold higher than WT controls. ACE2-MI mice exhibited a remarkable preservation of cardiac function and reduction of infarct size in comparison to WT-MI mice. Notably, ACE2 overexpression significantly reduced the MI-induced increase in apoptosis, macrophage infiltration, and HMGB1 and proinflammatory cytokine expression (TNF-α and IL-6). Moreover, in an in vitro study, ACE2 activation prevented the hypoxia-induced cell death and upregulation of HMGB1 in adult cardiomyocytes. This protective effect is correlated with downregulation of HMGB1 and downstream proinflammatory cascades, which could be useful for the development of novel treatment for ischemic heart disease.
Collapse
|
18
|
|
19
|
Hassanpour H, Afzali A, Fatemi Tabatabaie R, Torabi M, Alavi Y. Cardiac renin-angiotensin system (gene expression) and plasma angiotensin II in chickens with T3-induced pulmonary hypertension. Br Poult Sci 2016; 57:444-50. [DOI: 10.1080/00071668.2016.1162284] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- H. Hassanpour
- Department of Basic Sciences (Physiology Division), Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | - A. Afzali
- Department of Basic Sciences (Physiology Division), Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| | | | - M. Torabi
- Department of Physiology, Faculty of Veterinary Medicine, Shahid Chamran University, Ahvaz, Iran
| | - Y. Alavi
- Department of Basic Sciences (Physiology Division), Faculty of Veterinary Medicine, Shahrekord University, Shahrekord, Iran
| |
Collapse
|
20
|
Saha SA, Molnar J, Arora RR. Tissue ACE Inhibitors for Secondary Prevention of Cardiovascular Disease in Patients With Preserved Left Ventricular Function: A Pooled Meta-analysis of Randomized Placebo-controlled Trials. J Cardiovasc Pharmacol Ther 2016; 12:192-204. [PMID: 17875946 DOI: 10.1177/1074248407304791] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Objective. A pooled meta-analysis of published, randomized placebo-controlled clinical trials to evaluate the role of tissue angiotensin-converting enzyme (ACE) inhibitors in secondary prevention of cardiovascular disease in patients with preserved left ventricular function. Sources. Peer-reviewed journals listed in Index Medicus/MEDLINE, the Cochrane Central Register of Controlled Clinical Trials, and the Cochrane Database of Systematic Reviews. Study selection . Randomized placebo-controlled clinical trials of at least 12 months' duration, in patients with a prior cardiovascular event or at high risk for cardiovascular events, were analyzed. Data synthesis and analysis. A total of 31 555 patients (136 882 patient-years) from 4 trials were selected for the meta-analysis. Relative risk estimations were made using data pooled from these trials, and statistical significance was determined using the χ2 test. The number of patients needed to treat was also calculated for each outcome. Results. Tissue ACE inhibitors significantly reduced the risk of all-cause mortality, cardiovascular mortality, acute myocardial infarction, and stroke ( P < .001 for each). The need for invasive coronary revascularization was reduced ( P = .03), as was the risk of hospitalization for congestive heart failure ( P = .001). The occurrence of new-onset diabetes was also significantly reduced ( P < .001), but the risk of hospitalization for angina was not significantly affected ( P = .677). Treating about 100 patients for about 4.5 years would prevent 1 death, 1 non-fatal myocardial infarction, 1 cardiovascular death, or 1 invasive coronary revascularization. Conclusions. Tissue ACE inhibitors have demonstrated benefit when used for secondary prevention of cardiovascular disease in patients with preserved left ventricular function in randomized placebo-controlled clinical trials.
Collapse
Affiliation(s)
- Sandeep A Saha
- Department of Medicine, Chicago Medical School, North Chicago, Illinois 60064, USA
| | | | | |
Collapse
|
21
|
Bernardi S, Michelli A, Zuolo G, Candido R, Fabris B. Update on RAAS Modulation for the Treatment of Diabetic Cardiovascular Disease. J Diabetes Res 2016; 2016:8917578. [PMID: 27652272 PMCID: PMC5019930 DOI: 10.1155/2016/8917578] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 07/27/2016] [Indexed: 02/07/2023] Open
Abstract
Since the advent of insulin, the improvements in diabetes detection and the therapies to treat hyperglycemia have reduced the mortality of acute metabolic emergencies, such that today chronic complications are the major cause of morbidity and mortality among diabetic patients. More than half of the mortality that is seen in the diabetic population can be ascribed to cardiovascular disease (CVD), which includes not only myocardial infarction due to premature atherosclerosis but also diabetic cardiomyopathy. The importance of renin-angiotensin-aldosterone system (RAAS) antagonism in the prevention of diabetic CVD has demonstrated the key role that the RAAS plays in diabetic CVD onset and development. Today, ACE inhibitors and angiotensin II receptor blockers represent the first line therapy for primary and secondary CVD prevention in patients with diabetes. Recent research has uncovered new dimensions of the RAAS and, therefore, new potential therapeutic targets against diabetic CVD. Here we describe the timeline of paradigm shifts in RAAS understanding, how diabetes modifies the RAAS, and what new parts of the RAAS pathway could be targeted in order to achieve RAAS modulation against diabetic CVD.
Collapse
Affiliation(s)
- Stella Bernardi
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
- Division of Medicina Clinica, Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
- *Stella Bernardi:
| | - Andrea Michelli
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| | - Giulia Zuolo
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| | - Riccardo Candido
- Diabetes Centre, Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Via Puccini, 34100 Trieste, Italy
| | - Bruno Fabris
- Department of Medical Sciences, University of Trieste, Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
- Division of Medicina Clinica, Azienda Sanitaria Universitaria Integrata di Trieste (ASUITS), Cattinara Teaching Hospital, Strada di Fiume, 34100 Trieste, Italy
| |
Collapse
|
22
|
TCDD‑induced chick cardiotoxicity is abolished by a selective cyclooxygenase‑2 (COX‑2) inhibitor NS398. Arch Toxicol 2015; 88:1739-1748. [PMID: 24658325 DOI: 10.1007/s00204-014-1225-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2013] [Accepted: 03/10/2014] [Indexed: 01/05/2023]
Abstract
Halogenated aromatic hydrocarbons, including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), are known to cause severe heart defects in avian species. However, the mechanism of TCDD-induced chick cardiovascular toxicity is unclear. In this study, we investigated cyclooxygenase-2 (COX-2) as a possible mechanism of TCDD-induced cardiotoxicity. Fertile chicken eggs were injected with TCDD and a COX-2 selective inhibitor, NS398, and we investigated chick heart failure on day 10. We found that the chick heart to body weight ratio and atrial natriuretic factor mRNA expression were increased, but this increase was abolished with treatment of NS398. In addition, the morphological abnormality of an enlarged ventricle resulting from TCDD exposure was also abolished with co-treatment of TCDD and NS398. Our results suggested that TCDD-induced chick heart defects are mediated via the nongenomic pathway and that they do not require the genomic pathway.
Collapse
|
23
|
Červenka L, Melenovský V, Husková Z, Sporková A, Bürgelová M, Škaroupková P, Hwang SH, Hammock BD, Imig JD, Sadowski J. Inhibition of soluble epoxide hydrolase does not improve the course of congestive heart failure and the development of renal dysfunction in rats with volume overload induced by aorto-caval fistula. Physiol Res 2015; 64:857-73. [PMID: 26047375 DOI: 10.33549/physiolres.932977] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The detailed mechanisms determining the course of congestive heart failure (CHF) and associated renal dysfunction remain unclear. In a volume overload model of CHF induced by creation of aorto-caval fistula (ACF) in Hannover Sprague-Dawley (HanSD) rats we explored the putative pathogenetic contribution of epoxyeicosatrienoic acids (EETs), active products of CYP-450 dependent epoxygenase pathway of arachidonic acid metabolism, and compared it with the role of the renin-angiotensin system (RAS). Chronic treatment with cis-4-[4-(3-adamantan-1-yl-ureido) cyclohexyloxy]benzoic acid (c-AUCB, 3 mg/l in drinking water), an inhibitor of soluble epoxide hydrolase (sEH) which normally degrades EETs, increased intrarenal and myocardial EETs to levels observed in sham-operated HanSD rats, but did not improve the survival or renal function impairment. In contrast, chronic angiotensin-converting enzyme inhibition (ACEi, trandolapril, 6 mg/l in drinking water) increased renal blood flow, fractional sodium excretion and markedly improved survival, without affecting left ventricular structure and performance. Hence, renal dysfunction rather than cardiac remodeling determines long-term mortality in advanced stage of CHF due to volume overload. Strong protective actions of ACEi were associated with suppression of the vasoconstrictor/sodium retaining axis and activation of vasodilatory/natriuretic axis of the renin-angiotensin system in the circulating blood and kidney tissue.
Collapse
Affiliation(s)
- L Červenka
- Department of Pathophysiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Treskatsch S, Feldheiser A, Shaqura M, Dehe L, Habazettl H, Röpke TK, Shakibaei M, Schäfer M, Spies CD, Mousa SA. Cellular localization and adaptive changes of the cardiac delta opioid receptor system in an experimental model of heart failure in rats. Heart Vessels 2015; 31:241-50. [PMID: 25552382 DOI: 10.1007/s00380-014-0620-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Accepted: 12/19/2014] [Indexed: 01/30/2023]
Abstract
The role of the cardiac opioid system in congestive heart failure (CHF) is not fully understood. Therefore, this project investigated the cellular localization of delta opioid receptors (DOR) in left ventricle (LV) myocardium and adaptive changes in DOR and its endogenous ligand, the precursor peptide proenkephalin (PENK), during CHF. Following IRB approval, DOR localization was determined by radioligand binding using [H(3)]Naltrindole and by double immunofluorescence confocal analysis in the LV of male Wistar rats. Additionally, 28 days following an infrarenal aortocaval fistula (ACF) the extent of CHF and adaptions in left ventricular DOR and PENK expression were examined by hemodynamic measurements, RT-PCR, and Western blot. DOR specific membrane binding sites were identified in LV myocardium. DOR were colocalized with L-type Ca(2+)-channels (Cav1.2) as well as with intracellular ryanodine receptors (RyR) of the sarcoplasmatic reticulum. Following ACF severe congestive heart failure developed in all rats and was accompanied by up-regulation of DOR and PENK on mRNA as well as receptor proteins representing consecutive adaptations. These findings might suggest that the cardiac delta opioid system possesses the ability to play a regulatory role in the cardiomyocyte calcium homeostasis, especially in response to heart failure.
Collapse
Affiliation(s)
- Sascha Treskatsch
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Aarne Feldheiser
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Mohammed Shaqura
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Lukas Dehe
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Helmut Habazettl
- Institute of Physiology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Torsten K Röpke
- Department of Cardiology, Campus Charité Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Mehdi Shakibaei
- Institute of Anatomy, Ludwig-Maximilians-Universität München, Pettenkoferstraße 11, 80336, Munich, Germany.
| | - Michael Schäfer
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Claudia D Spies
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| | - Shaaban A Mousa
- Department of Anesthesiology and Intensive Care Medicine, Campus Charité Mitte and Campus Virchow Klinikum, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
25
|
Sdc1 overexpression inhibits the p38 MAPK pathway and lessens fibrotic ventricular remodeling in MI rats. Inflammation 2014; 36:603-15. [PMID: 23264165 DOI: 10.1007/s10753-012-9582-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Expression of the proteoglycan syndecan-1 (Sdc1) is increased in rats with myocardial infarction (MI). This study investigated the effects of Sdc1 overexpression on ventricular remodeling and cardiac function in MI and explored the possible mechanism through in vivo transfection of rats with recombinant adenovirus-carrying rat Sdc1 cDNA. Sprague-Dawley rats (n = 48) underwent intramyocardial injection in the marginal zone of the infarcted area immediately after ligation of the left anterior descending artery. The rats were divided into four groups according to the solution injected: MI Ad-GFP-Sdc1 transfection group, MI Ad-GFP control group, MI saline group, and sham operation group. Cardiac function and collagen expression of each group were examined, and the roles of inflammation, apoptosis, and p38 MAKP signal transduction pathway were investigated. Compared with the rats in the sham operation group, ventricular weight and collagen content increased in MI rats, and cardiac function declined. Substantial inflammatory cell infiltration was seen in the marginal zone of the infarction area, and a great number of myocardial cells were apoptotic. The p38 MAPK signaling pathway was clearly activated. Rats in the MI Ad-GFP-Sdc1 transfection group showed decreased ventricular weight, reduced collagen synthesis, and significant improvement of ventricular remodeling and cardiac function. Post-MI inflammatory cell infiltration and apoptosis was reduced, and the p38 MAPK signaling pathway was inhibited. Overexpression of Sdc1 can improve post-MI ventricular remodeling, and it is possible that the improvement is achieved through reducing apoptosis and suppressing inflammatory response and through the p38 MAPK signal transduction pathway.
Collapse
|
26
|
Dinicolantonio JJ, Lavie CJ, O'Keefe JH. Not all angiotensin-converting enzyme inhibitors are equal: focus on ramipril and perindopril. Postgrad Med 2013; 125:154-68. [PMID: 23933903 DOI: 10.3810/pgm.2013.07.2687] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Angiotensin-converting enzyme (ACE) inhibitors are a heterogeneous class, varying in pharmacologic properties, which have different therapeutic impacts on patient profiles, including lipophilicity, tissue-ACE binding, duration of action, half-life, and increased bradykinin availability. Among the ACE inhibitor class, the agent perindopril, in particular, has pleiotropic effects that are not equally shared by other ACE inhibitors, including bradykinin site selectivity and subsequent enhancement of nitric oxide and inhibition of endothelial cell apoptosis. Moreover, there is a large amount of evidence to suggest that perindopril therapy may reduce cardiovascular event rates in patients, yet perindopril is rarely prescribed in the United States. Ramipril is another ACE inhibitor with both a favorable clinical profile and impressive outcomes data. Our review compares the pharmacologic and trial data among perindopril, ramipril, and other ACE inhibitors. In patients with or at high risk for coronary heart disease who do not have heart failure, or in patients with heart failure with preserved ejection fraction, perindopril should be among the preferred treatment agents in the ACE inhibitor class. Ramipril has an impressive track record of improving cardiovascular outcomes, too, and should be considered a preferred agent among the ACE inhibitor class.
Collapse
|
27
|
Tamaki Y, Nakade Y, Yamauchi T, Makino Y, Yokohama S, Okada M, Aso K, Kanamori H, Ohashi T, Sato K, Nakao H, Haneda M, Yoneda M. Angiotensin II type 1 receptor antagonist prevents hepatic carcinoma in rats with nonalcoholic steatohepatitis. J Gastroenterol 2013; 48:491-503. [PMID: 22886508 DOI: 10.1007/s00535-012-0651-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2011] [Accepted: 07/23/2012] [Indexed: 02/07/2023]
Abstract
BACKGROUND Angiotensin II type 1 receptor blockers (ARBs) have been reported to attenuate hepatic fibrosis in non-alcoholic steatohepatitis (NASH). However, it is uncertain whether ARBs prevent hepatocarcinogenesis in NASH even after hepatic fibrosis has developed. METHODS Male Wistar rats were fed with a choline-deficient, L-amino acid-defined (CDAA) diet for 24 weeks, and then fed with the CDAA diet with telmisartan (2 mg/kg/day), a novel ARB, or vehicle for another 24 weeks. The liver histology and the expression of genes and proteins related to angiogenesis were investigated. RESULTS The 24-week CDAA diet induced liver cirrhosis. The 48-week CDAA diet exacerbated liver cirrhosis, and developed hepatocellular carcinoma (HCC) in 54.6 % of the rats concurrently with increases of hypoxia-inducible factor-1α (HIF-1α) protein and vascular endothelial growth factor (VEGF) mRNA, which are potent angiogenic factors in the liver. Telmisartan inhibited hepatic fibrosis and preneoplastic lesions and prevented the development of HCC along with inducing decreases in HIF-1α protein and VEGF mRNA. CONCLUSIONS These data indicated that telmisartan may prevent hepatocarcinogenesis through the inhibition of hepatic angiogenesis even after liver cirrhosis has been established.
Collapse
MESH Headings
- Amino Acids/administration & dosage
- Angiotensin II Type 1 Receptor Blockers/therapeutic use
- Animals
- Anticarcinogenic Agents/therapeutic use
- Benzimidazoles/therapeutic use
- Benzoates/therapeutic use
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/prevention & control
- Cell Transformation, Neoplastic/drug effects
- Choline Deficiency/complications
- Diet/adverse effects
- Drug Evaluation, Preclinical/methods
- Fatty Liver/complications
- Fatty Liver/drug therapy
- Fatty Liver/metabolism
- Fatty Liver/pathology
- Gene Expression Regulation/drug effects
- Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Liver/blood supply
- Liver Cirrhosis/complications
- Liver Cirrhosis/drug therapy
- Liver Cirrhosis/metabolism
- Liver Cirrhosis/pathology
- Liver Neoplasms, Experimental/etiology
- Liver Neoplasms, Experimental/prevention & control
- Male
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/prevention & control
- Non-alcoholic Fatty Liver Disease
- Rats
- Rats, Wistar
- Telmisartan
- Vascular Endothelial Growth Factor A/biosynthesis
- Vascular Endothelial Growth Factor A/genetics
Collapse
Affiliation(s)
- Yosui Tamaki
- Division of Metabolism and Biosystemic Science, Department of Internal Medicine, Asahikawa Medical College, Asahikawa, Hokkaido, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Shi W, Meszaros JG, Zeng SJ, Sun YY, Zuo MX. Living high training low induces physiological cardiac hypertrophy accompanied by down-regulation and redistribution of the renin-angiotensin system. Acta Pharmacol Sin 2013; 34:342-51. [PMID: 23377552 PMCID: PMC4002499 DOI: 10.1038/aps.2012.210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 11/28/2012] [Indexed: 01/13/2023]
Abstract
AIM Living high training low" (LHTL) is an exercise-training protocol that refers living in hypoxia stress and training at normal level of O2. In this study, we investigated whether LHTL caused physiological heart hypertrophy accompanied by changes of biomarkers in renin-angiotensin system in rats. METHODS Adult male SD rats were randomly assigned into 4 groups, and trained on living low-sedentary (LLS, control), living low-training low (LLTL), living high-sedentary (LHS) and living high-training low (LHTL) protocols, respectively, for 4 weeks. Hematological parameters, hemodynamic measurement, heart hypertrophy and plasma angiotensin II (Ang II) level of the rats were measured. The gene and protein expression of angiotensin-converting enzyme (ACE), angiotensinogen (AGT) and angiotensin II receptor I (AT1) in heart tissue was assessed using RT-PCR and immunohistochemistry, respectively. RESULTS LLTL, LHS and LHTL significantly improved cardiac function, increased hemoglobin concentration and RBC. At the molecular level, LLTL, LHS and LHTL significantly decreased the expression of ACE, AGT and AT1 genes, but increased the expression of ACE and AT1 proteins in heart tissue. Moreover, ACE and AT1 protein expression was significantly increased in the endocardium, but unchanged in the epicardium. CONCLUSION LHTL training protocol suppresses ACE, AGT and AT1 gene expression in heart tissue, but increases ACE and AT1 protein expression specifically in the endocardium, suggesting that the physiological heart hypertrophy induced by LHTL is regulated by region-specific expression of renin-angiotensin system components.
Collapse
Affiliation(s)
- Wei Shi
- Laboratory of Neuroscience and Brain Development, School of Life Science, Beijing Normal University, Beijing 100875, China
- School of Biomedical Sciences, Kent State University, Kent, OH 44240, USA
| | - J Gary Meszaros
- Department of Integrative Medical Sciences, Northeastern Ohio Universities Colleges of Medicine and Pharmacy, Rootstown, OH 44272, USA
| | - Shao-ju Zeng
- Laboratory of Neuroscience and Brain Development, School of Life Science, Beijing Normal University, Beijing 100875, China
| | - Ying-yu Sun
- Laboratory of Neuroscience and Brain Development, School of Life Science, Beijing Normal University, Beijing 100875, China
| | - Ming-xue Zuo
- Laboratory of Neuroscience and Brain Development, School of Life Science, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
29
|
Abstract
The RAS (renin-angiotensin system) is one of the earliest and most extensively studied hormonal systems. The RAS is an atypical hormonal system in several ways. The major bioactive peptide of the system, AngII (angiotensin II), is neither synthesized in nor targets one specific organ. New research has identified additional peptides with important physiological and pathological roles. More peptides also mean newer enzymatic cascades that generate these peptides and more receptors that mediate their function. In addition, completely different roles of components that constitute the RAS have been uncovered, such as that for prorenin via the prorenin receptor. Complexity of the RAS is enhanced further by the presence of sub-systems in tissues, which act in an autocrine/paracrine manner independent of the endocrine system. The RAS seems relevant at the cellular level, wherein individual cells have a complete system, termed the intracellular RAS. Thus, from cells to tissues to the entire organism, the RAS exhibits continuity while maintaining independent control at different levels. The intracellular RAS is a relatively new concept for the RAS. The present review provides a synopsis of the literature on this system in different tissues.
Collapse
|
30
|
Angiotensin-Converting Enzyme 2 (ACE2) Is a Key Modulator of the Renin Angiotensin System in Health and Disease. INTERNATIONAL JOURNAL OF PEPTIDES 2012; 2012:256294. [PMID: 22536270 PMCID: PMC3321295 DOI: 10.1155/2012/256294] [Citation(s) in RCA: 399] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/05/2011] [Indexed: 12/29/2022]
Abstract
Angiotensin-converting enzyme 2 (ACE2) shares some homology with angiotensin-converting enzyme (ACE) but is not inhibited by ACE inhibitors. The main role of ACE2 is the degradation of Ang II resulting in the formation of angiotensin 1–7 (Ang 1–7) which opposes the actions of Ang II. Increased Ang II levels are thought to upregulate ACE2 activity, and in ACE2 deficient mice Ang II levels are approximately double that of wild-type mice, whilst Ang 1–7 levels are almost undetectable. Thus, ACE2 plays a crucial role in the RAS because it opposes the actions of Ang II. Consequently, it has a beneficial role in many diseases such as hypertension, diabetes, and cardiovascular disease where its expression is decreased. Not surprisingly, current therapeutic strategies for ACE2 involve augmenting its expression using ACE2 adenoviruses, recombinant ACE2 or compounds in these diseases thereby affording some organ protection.
Collapse
|
31
|
Abstract
Previously, we demonstrated that angiotensin II type 2 (AT(2)) receptors have a role in natriuresis in obese Zucker rats (OZR). In the present study, we investigated the role of a novel, non-peptide agonist, C21, in natriuresis via AT(2) receptor activation in OZR. Infusion of C21 (1 and 5 μg kg(-1) min(-1)) into rats under anesthesia caused a dose-dependent increase in urine flow (UF) and urinary Na volume (U(Na)V). These effects of C21 were blocked by pre-infusion of the AT(2) receptor antagonist, PD123319, (50 μg kg(-1) min(-1)), suggesting involvement of the AT(2) receptor. Infusion of C21 (5 μg kg(-1) min(-1)) significantly increased the fractional excretion of sodium without changing the glomerular filtration rate or blood pressure, suggesting a tubular effect. Similarly, C21 infusion increased the fractional excretion of lithium, suggesting a proximal tubular effect. Furthermore, we tested the effect of C21 on natriuresis after blocking two main, distal-nephron Na transporters, the epithelial Na channels (ENaC), with amiloride (AM, 3 mg kg(-1) body wt), and the NaCl cotransporters (NCC), with bendroflumethiazide (BFTZ, 7 mg kg(-1) body wt). Infusion of AM + BFTZ caused significant increases in both diuresis and natriuresis, which were further increased by infusion of C21 (5 μg kg(-1) min(-1)). Natriuresis in response to C21 was associated with increases in urinary NO and cGMP levels. The data indicate that the AT(2) receptor agonist, C21, promotes natriuresis via AT(2) receptor activation and that this effect is potentially based in the proximal tubules and linked to the nitric oxide/cyclic guanosine monophosphate pathway. The natriuretic response to C21 may have therapeutic significance by improving kidney function in obesity.
Collapse
|
32
|
Lei J, Xue SN, Wu W, Zhou SX, Zhang YL, Yuan GY, Wang JF. Increased level of soluble syndecan-1 in serum correlates with myocardial expression in a rat model of myocardial infarction. Mol Cell Biochem 2011; 359:177-82. [DOI: 10.1007/s11010-011-1012-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2011] [Accepted: 07/27/2011] [Indexed: 10/17/2022]
|
33
|
Angiotensin-converting enzyme 2 is a key modulator of the renin-angiotensin system in cardiovascular and renal disease. Curr Opin Nephrol Hypertens 2011; 20:62-8. [PMID: 21099686 DOI: 10.1097/mnh.0b013e328341164a] [Citation(s) in RCA: 119] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Angiotensin-converting enzyme 2 (ACE2) has recently emerged as a key regulator of the renin-angiotensin system in both health and disease. RECENT FINDINGS ACE2 deficiency is associated with elevated tissue and circulating levels of angiotensin II and reduced levels of angiotensin 1-7. Phenotypically, this results in a modest elevation in systolic blood pressure and left ventricular hypertrophy. In atherosclerosis-prone apolipoprotein E knockout mice, ACE2 deficiency results in augmented vascular inflammation and an inflammatory response that contributes to increased atherosclerotic plaque formation. In the kidney, ACE2 deficiency is associated with progressive glomerulosclerosis. Interventions such as ACE2 replenishment or augmentation of its actions have proven successful in reducing hypertension, plaque accumulation, and renal and cardiac damage in a range of different models. Although promising, the balance of the renin-angiotensin system remains complicated, with some evidence that overexpression of ACE2 may have adverse cardiac effects, and ACE2 and its metabolic products may promote epithelial-to-mesenchymal transition. SUMMARY Repletion of ACE2's activities offers a new strategy to complement current clinical interventions in treating hypertension, renal and cardiovascular disease. In particular conditions where ACE inhibition and angiotensin receptor blockade are partially effective, the adjunctive actions of ACE2 may not only reduce clinical escape but also augment the efficacy of interventions.
Collapse
|
34
|
Response to angiotensin-converting enzyme inhibition is selectively blunted by high sodium in angiotensin-converting enzyme DD genotype: evidence for gene-environment interaction in healthy volunteers. J Hypertens 2011; 28:2414-21. [PMID: 20829712 DOI: 10.1097/hjh.0b013e32833eaf34] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Renin-angiotensin-aldosterone system blockade is a cornerstone in cardiovascular protection. Angiotensin-converting enzyme (ACE)-DD genotype has been associated with resistance to angiotensin-converting enzyme inhibition (ACEi), but data are conflicting. As sodium intake modifies the effect of ACEi as well as the genotype-phenotype relationship, we hypothesize gene-environment interaction between sodium-status, the response to ACEi, and ACE genotype. METHOD Thirty-five male volunteers (26 ± 9 years; II n = 6, ID n = 18, DD n = 11) were studied during placebo and ACEi (double blind, enalapril 20 mg/day) on low [7 days 50 mmol Na/day (low salt)] and high [7 days 200 mmol Na/day (high salt)] sodium, with a washout of 6 weeks in-between. After each period mean arterial pressure (MAP) was measured before and during graded infusion of angiotensin II (Ang II). RESULTS During high salt, ACEi reduced MAP in II and ID, but not in DD [II: 88 (78-94) versus 76 (72-88); ID: 87 (84-91) versus 83 (79-87); both P < 0.05 and DD: 86 (82-96) versus 88 (80-90); ns, P < 0.05 between genotypes]. However, during low salt, ACEi reduced MAP in all genotype groups [II: 83 (78-89) versus 77 (72-83); ID: 88 (84-91) versus 82 (78-86); DD: 84 (80-91) versus 81 (75-85); all P < 0.05]. During high salt + ACEi, the Ang II response was blunted in DD, with an 18% rise in MAP during the highest dose versus 22 and 31% in ID and II (P < 0.05). Low salt annihilated these differences. CONCLUSION In healthy participants, the MAP response to ACEi is selectively blunted in DD genotype during high salt, accompanied by blunted sensitivity to Ang II. Low salt corrects both abnormalities. Further analysis of this gene-environment interaction in patients may contribute to strategies for improvement of individual treatment efficacy.
Collapse
|
35
|
Aortocaval fistula in rat: a unique model of volume-overload congestive heart failure and cardiac hypertrophy. J Biomed Biotechnol 2011; 2011:729497. [PMID: 21274403 PMCID: PMC3025398 DOI: 10.1155/2011/729497] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2010] [Accepted: 12/09/2010] [Indexed: 11/17/2022] Open
Abstract
Despite continuous progress in our understanding of the pathogenesis of congestive heart failure (CHF) and its management, mortality remains high. Therefore, development of reliable experimental models of CHF and cardiac hypertrophy is essential to better understand disease progression and allow new therapy developement. The aortocaval fistula (ACF) model, first described in dogs almost a century ago, has been adopted in rodents by several groups including ours. Although considered to be a model of high-output heart failure, its long-term renal and cardiac manifestations are similar to those seen in patients with low-output CHF. These include Na+-retention, cardiac hypertrophy and increased activity of both vasoconstrictor/antinatriureticneurohormonal systems and compensatory vasodilating/natriuretic systems. Previous data from our group and others suggest that progression of cardiorenal pathophysiology in this model is largely determined by balance between opposing hormonal forces, as reflected in states of CHF decompensation that are characterized by overactivation of vasoconstrictive/Na+-retaining systems. Thus, ACF serves as a simple, cheap, and reproducible platform to investigate the pathogenesis of CHF and to examine efficacy of new therapeutic approaches. Hereby, we will focus on the neurohormonal, renal, and cardiac manifestations of the ACF model in rats, with special emphasis on our own experience.
Collapse
|
36
|
Hirose T, Mori N, Totsune K, Morimoto R, Maejima T, Kawamura T, Metoki H, Asayama K, Kikuya M, Ohkubo T, Kohzuki M, Takahashi K, Imai Y. Gene expression of (pro)renin receptor is upregulated in hearts and kidneys of rats with congestive heart failure. Peptides 2009; 30:2316-22. [PMID: 19765626 DOI: 10.1016/j.peptides.2009.09.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2009] [Revised: 09/10/2009] [Accepted: 09/10/2009] [Indexed: 12/23/2022]
Abstract
Recent studies have revealed that (pro)renin receptor ((P)RR), a newly identified member of the renin-angiotensin system, was associated with organ damage in the kidney. However, there has been little information for (P)RR in hearts. To investigate the regulation of (P)RR in heart failure, we examined the expression of (P)RR in hearts and kidneys of rats with congestive heart failure (CHF) due to coronary ligation by quantitative RT-PCR and immunohistochemistry. Significantly increased levels of (P)RR mRNA were found in the atrium, right ventricle, non-infarcted part of left ventricle, infarcted part of left ventricle and kidney of CHF rats, when compared with sham operated rats (about 1.6-fold, 1.4-fold, 1.6-fold, 1.7-fold and 1.5-fold, respectively). Expression levels of mRNAs encoding renin and angiotensinogen in these heart and kidney tissues were also increased in the CHF rats. Immunohistochemistry showed positive (P)RR immunostaining in the myocardium, the renal tubular cells, and vascular smooth muscle and endothelial cells in the heart and the kidney. The renal tubular cells were more intensely immunostained in CHF rats than in sham operated rats. These findings suggest that the expression of (P)RR is increased in the hearts and kidneys of rats with heart failure, and that (P)RR may contribute to heart failure.
Collapse
Affiliation(s)
- Takuo Hirose
- Department of Clinical Pharmacology and Therapeutics, Tohoku University Graduate School of Pharmaceutical Sciences and Medicine, Sendai 980-8578, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Borin TF, Miyakawa AA, Cardoso L, de Figueiredo Borges L, Gonçalves GA, Krieger JE. Apoptosis, cell proliferation and modulation of cyclin-dependent kinase inhibitor p21(cip1) in vascular remodelling during vein arterialization in the rat. Int J Exp Pathol 2009; 90:328-37. [PMID: 19563615 DOI: 10.1111/j.1365-2613.2009.00648.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Neo-intima development and atherosclerosis limit long-term vein graft use for revascularization of ischaemic tissues. Using a rat model, which is technically less challenging than smaller rodents, we provide evidence that the temporal morphological, cellular, and key molecular events during vein arterialization resemble the human vein graft adaptation. Right jugular vein was surgically connected to carotid artery and observed up to 90 days. Morphometry demonstrated gradual thickening of the medial layer and important formation of neo-intima with deposition of smooth muscle cells (SMC) in the subendothelial layer from day 7 onwards. Transmission electron microscopy showed that SMCs switch from the contractile to synthetic phenotype on day 3 and new elastic lamellae formation occurs from day 7 onwards. Apoptosis markedly increased on day 1, while alpha-actin immunostaining for SMC almost disappeared by day 3. On day 7, cell proliferation reached the highest level and cellular density gradually increased until day 90. The relative magnitude of cellular changes was higher in the intima vs. the media layer (100 vs. 2 times respectively). Cyclin-dependent kinase inhibitors (CDKIs) p27(Kip1) and p16(INKA) remained unchanged, whereas p21(Cip1) was gradually downregulated, reaching the lowest levels by day 7 until day 90. Taken together, these data indicate for the first time that p21(Cip1) is the main CDKI protein modulated during the arterialization process the rat model of vein arterialization that may be useful to identify and validate new targets and interventions to improve the long-term patency of vein grafts.
Collapse
Affiliation(s)
- Thaiz Ferraz Borin
- Laboratory of Genetic and Molecular Cardiology/LIM-13, Heart Institute (InCor), University of Sao Paulo Medical School, Sao Paulo, SP, Brazil
| | | | | | | | | | | |
Collapse
|
38
|
Radi ZA, Murad Y. Cellular expression of renal, cardiac and pulmonary inducible nitric oxide synthase in double-transgenic mice expressing human renin and angiotensinogen genes. Clin Exp Pharmacol Physiol 2008; 36:571-5. [PMID: 19673942 DOI: 10.1111/j.1440-1681.2008.05120.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
1. Hypertensive mice expressing the human renin (REN) and angiotensinogen (AGT) genes are used as a model for human hypertension. 2. The aim of the present study was to investigate the cellular expression and distribution of inducible nitric oxide synthase (iNOS) using immunohistochemistry in lung, heart and kidney tissues from a model of human hypertension using male and female double-transgenic (h-Ang 204/1h-Ren6) mice and wild-type C57/BI6J mice as controls. 3. In the kidney, the pattern of iNOS expression in various renal microanatomical regions during hypertension was similar to that of age-matched controls, except in the medullary ascending limb (MAL). In hypertension, iNOS expression was downregulated in the MAL. No significant differences in iNOS expression were seen between control or hypertensive mice in various cardiac microanatomical locations. In the lungs of hypertensive mice, iNOS expression was upregulated in bronchial airway epithelium and bronchial and vascular smooth muscle cells, but downregulated in alveolar macrophages, alveolar septa and pulmonary vascular endothelial cells. Expression of iNOS was similar between male and female mice in the kidney, heart and lungs. 4. In conclusion, iNOS regulation in hypertension is complex and depends on the cell type in which it is expressed and the localization of the cell type in the cardiorenal and pulmonary systems.
Collapse
Affiliation(s)
- Zaher A Radi
- Drug Safety Research and Development, Pfizer Global Research and Development, St Louis, Misouri 63017, USA.
| | | |
Collapse
|
39
|
Radi ZA, Ostroski R. Pulmonary and cardiorenal cyclooxygenase-1 (COX-1), -2 (COX-2), and microsomal prostaglandin E synthase-1 (mPGES-1) and -2 (mPGES-2) expression in a hypertension model. Mediators Inflamm 2008; 2007:85091. [PMID: 17641732 PMCID: PMC1906712 DOI: 10.1155/2007/85091] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Accepted: 03/16/2007] [Indexed: 11/18/2022] Open
Abstract
Hypertensive mice that express the human renin and angiotensinogen genes are used as a model for human hypertension because they develop hypertension secondary to increased renin-angiotensin system activity. Our study investigated the cellular localization and distribution of COX-1, COX-2, mPGES-1, and mPGES-2 in organ tissues from a mouse model of human hypertension. Male (n = 15) and female (n = 15) double transgenic mice (h-Ang 204/1 h-Ren 9) were used in the study. Lung, kidney, and heart tissues were obtained from mice at necropsy and fixed in 10% neutral buffered formalin followed by embedding in paraffin wax. Cut sections were stained immunohistochemically with antibodies to COX-1, COX-2, mPGES-1, and mPGES-2 and analyzed by light microscopy. Renal expression of COX-1 was the highest in the distal convoluted tubules, cortical collecting ducts, and medullary collecting ducts; while proximal convoluted tubules lacked COX-1 expression. Bronchial and bronchiolar epithelial cells, alveolar macrophages, and cardiac vascular endothelial cells also had strong COX-1 expression, with other renal, pulmonary, or cardiac microanatomic locations having mild-to-moderate expression. mPGES-2 expression was strong in the bronchial and bronchiolar epithelial cells, mild to moderate in various renal microanatomic locations, and absent in cardiac tissues. COX-2 expression was strong in the proximal and distal convoluted tubules, alveolar macrophages, and bronchial and bronchiolar epithelial cells. Marked mPGES-1 was present only in bronchial and bronchiolar epithelial cells; while mild-to-moderate expression was present in other pulmonary, renal, or cardiac microanatomic locations. Expression of these molecules was similar between males and females. Our work suggests that in hypertensive mice, there are (a) significant microanatomic variations in the pulmonary, renal, and cardiac distribution and cellular localization of COX-1, COX-2, mPGES-1, and mPGES-2, and (b) no differences in expression between genders.
Collapse
Affiliation(s)
- Zaher A. Radi
- Drug Safety Research & Development, Pfizer Global Research and Development, 2800 Plymouth Road, Building 50-G0503,
Ann Arbor, MI 48105, USA
- *Zaher A. Radi:
| | - Robert Ostroski
- Department of Cardiovascular Pharmacology, Pfizer Global Research and Development, 2800 Plymouth Road,
Building 50-G0503, Ann Arbor, MI 48105, USA
| |
Collapse
|
40
|
Thompson J, Raitt J, Hutchings L, Drenos F, Bjargo E, Loset A, Grocott M, Montgomery H. Angiotensin-converting enzyme genotype and successful ascent to extreme high altitude. High Alt Med Biol 2008; 8:278-85. [PMID: 18081503 DOI: 10.1089/ham.2007.1044] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Interindividual variation in acclimatization to altitude suggests a genetic component, and several candidate genes have been proposed. One such candidate is a polymorphism in the angiotensin converting enzyme (ACE) gene, where the insertion (I-allele), rather than the deletion (D-allele), of a 287 base pair sequence has been associated with lower circulating and tissue ACE activity and has a greater than normal frequency among elite endurance athletes and, in a single study, among elite high altitude mountaineers. We tested the hypothesis that the I-allele is associated with successful ascent to the extreme high altitude of 8000 m. 141 mountaineers who had participated in expeditions attempting to climb an 8000-m peak completed a questionnaire and provided a buccal swab for ACE I/D genotyping. ACE genotype was determined in 139 mountaineers. ACE genotype distribution differed significantly between those who had successfully climbed beyond 8000 m and those who had not (p = 0.003), with a relative overrepresentation of the I-allele among the successful group (0.55 vs. 0.36 in successful vs. unsuccessful, respectively). The I-allele was associated with increased maximum altitudes achieved: 8079 +/- 947 m for DDs, 8107 +/- 653 m for IDs, and 8559 +/- 565 m for IIs (p = 0.007). There was no statistical difference in ACE genotype frequency between those who climbed to over 8000 m using supplementary oxygen and those who did not (p = 0.267). This study demonstrates an association between the ACE I-allele and successful ascent to over 8000 m.
Collapse
Affiliation(s)
- Julian Thompson
- UCL Institute for Human Health and Performance, London, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Ferreira JCB, Bacurau AV, Evangelista FS, Coelho MA, Oliveira EM, Casarini DE, Krieger JE, Brum PC. The role of local and systemic renin angiotensin system activation in a genetic model of sympathetic hyperactivity-induced heart failure in mice. Am J Physiol Regul Integr Comp Physiol 2008; 294:R26-32. [DOI: 10.1152/ajpregu.00424.2007] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sympathetic hyperactivity (SH) and renin angiotensin system (RAS) activation are commonly associated with heart failure (HF), even though the relative contribution of these factors to the cardiac derangement is less understood. The role of SH on RAS components and its consequences for the HF were investigated in mice lacking α2Aand α2Cadrenoceptor knockout (α2A/α2CARKO) that present SH with evidence of HF by 7 mo of age. Cardiac and systemic RAS components and plasma norepinephrine (PN) levels were evaluated in male adult mice at 3 and 7 mo of age. In addition, cardiac morphometric analysis, collagen content, exercise tolerance, and hemodynamic assessments were made. At 3 mo, α2A/α2CARKO mice showed no signs of HF, while displaying elevated PN, activation of local and systemic RAS components, and increased cardiomyocyte width (16%) compared with wild-type mice (WT). In contrast, at 7 mo, α2A/α2CARKO mice presented clear signs of HF accompanied only by cardiac activation of angiotensinogen and ANG II levels and increased collagen content (twofold). Consistent with this local activation of RAS, 8 wk of ANG II AT1receptor blocker treatment restored cardiac structure and function comparable to the WT. Collectively, these data provide direct evidence that cardiac RAS activation plays a major role underlying the structural and functional abnormalities associated with a genetic SH-induced HF in mice.
Collapse
|
42
|
Abstract
Cardiac mast cells proliferate in cardiovascular diseases. In myocardial ischemia, mast cell mediators contribute to coronary vasoconstriction, arrhythmias, leukocyte recruitment, and tissue injury and repair. Arrhythmic dysfunction, coronary vasoconstriction, and contractile failure are also characteristic of cardiac anaphylaxis. In coronary atherosclerosis, mast cell mediators facilitate cholesterol accumulation and plaque destabilization. In cardiac failure, mast cell chymase causes myocyte apoptosis and fibroblast proliferation, leading to ventricular dysfunction. Chymase and tryptase also contribute to fibrosis in cardiomyopathies and myocarditis. In addition, mast cell tumor necrosis factor-alpha promotes myocardial remodeling. Cardiac remodeling and hypertrophy in end-stage hypertension are also induced by mast cell mediators and proteases. We recently discovered that cardiac mast cells contain and release renin, which initiates local angiotensin formation. Angiotensin causes coronary vasoconstriction, arrhythmias, fibrosis, apoptosis, and endothelin release, all demonstrated mechanisms of mast-cell-associated cardiac disease. The effects of angiotensin are further amplified by the release of norepinephrine from cardiac sympathetic nerves. Our discovery of renin in cardiac mast cells and its release in pathophysiological conditions uncovers an important new pathway in the development of mast-cell-associated heart diseases. Several steps in this novel pathway may constitute future therapeutic targets.
Collapse
Affiliation(s)
- Alicia C Reid
- Department of Physiology and Biophysics, Weill Cornell Medical College, New York, NY 10021, USA
| | | | | |
Collapse
|
43
|
Fujii Y, Yamane T, Orito K, Osamura K, Wakao Y. Increased chymase-like activity in a dog with congenital pulmonic stenosis. J Vet Cardiol 2007; 9:39-42. [DOI: 10.1016/j.jvc.2005.07.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2005] [Revised: 07/24/2005] [Accepted: 07/29/2005] [Indexed: 10/23/2022]
|
44
|
Cheng G, Xu G, Cai HW, Wang HH, Bao XF. Effect of atorvastatin on non-ischemic heart failure and matrix metalloproteinase-2 and 9 in rats. Acta Pharmacol Sin 2007; 28:511-7. [PMID: 17376290 DOI: 10.1111/j.1745-7254.2007.00515.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIM To examine the role of atorvastatin on volume-overload-induced heart failure and to test the hypothesis that atorvastatin inhibits MMP-2 and 9 expression in heart failure with non-ischemic etiology. METHODS Arteriovenous (AV) fistula-treated rats were administered with atorvastatin (3 mg/kg/d) or vehicle for 17 weeks. Ventricular hypertrophy and heart failure were assessed by echocardiography, B-type natriuretic peptide BNP mRNA level and morphological measurement. MMP-2, 9 expression were measured by Western blot and zymography. RESULTS Atorvastatin decreased left ventricular end diastolic diameter from 6.86+/-0.51 mm to 6.28+/-0.37 mm (P<0.05), increased fractioning shortening from 41.4%+/-4.5% to 52.7%+/-4.2% (P<0.01), decreased ratio of BNP/GAPDH mRNA level from 0.43+/-0.03 to 0.27+/-0.03 (P<0.05). Similar data were observed for morphological measurement. Protein expression and enzyme activity of MMP-2 and 9 in the left ventricle tissue were significantly increased 18 weeks after surgery and atorvastatin also prevented those changes. CONCLUSION Left ventricular remodeling induced by AV fistula was profoundly changed by atorvastatin treatment. Hypertrophy was attenuated and global function was improved. These positive effects of atorvastatin on heart failure were associated with decreased MMP-2 and 9 protein expression and enzyme activity.
Collapse
Affiliation(s)
- Gang Cheng
- Department of Cardiology, the Second Affiliated Hospital, Zhejiang University, Medical School, Hangzhou 310009, China
| | | | | | | | | |
Collapse
|
45
|
Spencer TA, Smyth SH, Wittich G, Hunter GC. Delayed presentation of traumatic aortocaval fistula: A report of two cases and a review of the associated compensatory hemodynamic and structural changes. J Vasc Surg 2006; 43:836-40. [PMID: 16616246 DOI: 10.1016/j.jvs.2005.12.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Accepted: 12/01/2005] [Indexed: 11/16/2022]
Abstract
Chronic aortocaval fistula (ACF) is a rare complication of gunshot wounds to the abdomen. Herein we report two cases of traumatic ACF: one asymptomatic and the other presenting with congestive heart failure (CHF) 20 and 30 years, respectively, after their initial injury. The recent onset of CHF, the presence of a continuous abdominal bruit, and, in the second patient, a history of penetrating trauma suggested the diagnosis of ACF. The diagnosis was confirmed by computed tomography scanning in both patients. Surgical repair of the ACF in the symptomatic patient resulted in resolution of the CHF and reversed the dilatation of the aorta and inferior vena cava. The asymptomatic patient was lost to follow-up. CHF in a young male patient with a history of penetrating abdominal trauma should alert the surgeon to this rare complication.
Collapse
Affiliation(s)
- Todd A Spencer
- Department of Surgery, The University of Texas Medical Branch, Galveston 77555-0541, USA
| | | | | | | |
Collapse
|
46
|
Karram T, Hoffman A, Bishara B, Brodsky S, Golomb E, Winaver J, Abassi Z. Induction of cardiac hypertrophy by a controlled reproducible sutureless aortocaval shunt in the mouse. J INVEST SURG 2006; 18:325-34. [PMID: 16319054 DOI: 10.1080/08941930500328839] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Much of the understanding about the pathophysiological responses to chronic cardiac overload has been gained by the use of rat and dog models of aortocaval fistula (ACF). The use of a similar model in genetically manipulated mice may further elucidate the molecular mechanisms in these responses. The only reports about ACF in mice to date have applied a needle puncture to create the ACF, which may result in an uncontrolled and irreproducible size of the shunt, and require several weeks to induce the characteristic cardiac changes. In order to obtain a more consistent approach to characterize this mode of cardiac hyperfunction, we present a surgical murine model of ACF that results in rapid progression of the typical systemic and cardiac changes. A sutureless side-to-side infrarenal surgical anastomosis of 0.6-0.8 mm in diameter was created between the abdominal aorta and inferior vena cava in ICR (Institute of Cancer Research) mice. Six to 7 days later, significant cardiac hypertrophy developed. The heart/body weight ratio increased from 0.45 +/- 0.02% in control mice to 0.77 +/- 0.03% in mice with ACF (p < .003). The dry heart weight ratio increased from 0.099 +/- 0.0033% to 0.13 +/- 0.008% (p < .006). The ACF dramatically induced the atrial and ventricular expression of atrial natriuretic factor mRNA, and increased the total cardiac content of endothelin-1 (162.5 +/- 50.6 vs. 83.9 +/- 9.0 pg). Mean arterial pressure in anesthetized mice with ACF decreased from 69.8 +/- 4.9 to 54.8 +/- 5.5 mm Hg (p < .025). Urinary sodium excretion returned to preoperative levels several days following surgery. These results demonstrate that cardiac hypertrophy could be rapidly and reproducibly achieved in mice by the placement of a surgical ACF. This model, when applied in genetically manipulated mice, may be a valuable tool for functional genomic studies about the pathogenesis of cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Tony Karram
- Department of Vascular Surgery and Transplantation Unit, Rambam Medical Center, Haifa, Israel
| | | | | | | | | | | | | |
Collapse
|
47
|
Seckin D, Ilhan N, Ilhan N, Ozbay Y. The relationship between ACE insertion/deletion polymorphism and coronary artery disease with or without myocardial infarction. Clin Biochem 2006; 39:50-4. [PMID: 16303122 DOI: 10.1016/j.clinbiochem.2005.10.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2005] [Revised: 07/18/2005] [Accepted: 10/17/2005] [Indexed: 11/27/2022]
Abstract
OBJECTIVES Presence of the D allele or homozygosity for the deletion (D) allele of the angiotensicen-converting enzyme (ACE) insertion/deletion (I/D) polymorphism has been discussed as potent risk factor for coronary artery disease (CAD) and myocardial infarction (MI). The D allele is associated with higher levels of circulating ACE and therefore may predispose one to cardiovascular damage. DESIGN AND METHODS The study presented here was performed to investigate the association between the ACE genotype and ACE levels. The study group was comprised of 118 angiographically verified CAD patients. 65 patients were MI (+) and 53 patients were MI (-) in this group. A total of 70 healthy individuals were taken as controls. Genomic DNA of 188 subjects was extracted from whole blood. The polymerase chain reaction was used for ACE genotyping, and ACE levels were measured by ELISA. RESULTS The D allele was found to be significantly more frequent in patients with MI (+) compared with controls (P = 0.024). ACE levels were significantly higher in both MI (-) and MI (+) groups with CAD patients than in controls (P < 0.005). Plasma ACE level was higher in all three groups in the DD genotype compared to II genotype. In groups I and III, this was statistically significant (P < 0.0001, P < 0.01). CONCLUSIONS It was shown that the I/D polymorphism in the gene for ACE is a genetic risk factor for CAD patients who have a history of MI. ACE insertion/deletion gene polymorphism is also associated with plasma ACE levels in CAD patients with a history of MI.
Collapse
Affiliation(s)
- Dilara Seckin
- Department of Biochemistry, Firat University Firat Medical Center, Elazig, Turkey.
| | | | | | | |
Collapse
|
48
|
Saavedra JM, Ando H, Armando I, Baiardi G, Bregonzio C, Juorio A, Macova M. Anti-stress and anti-anxiety effects of centrally acting angiotensin II AT1 receptor antagonists. ACTA ACUST UNITED AC 2005; 128:227-38. [PMID: 15837532 DOI: 10.1016/j.regpep.2004.12.015] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The brain and the peripheral (hormonal) angiotensin II systems are stimulated during stress. Activation of brain angiotensin II AT(1) receptors is required for the stress-induced hormone secretion, including CRH, ACTH, corticoids and vasopressin, and for stimulation of the central sympathetic activity. Long-term peripheral administration of the angiotensin II AT(1) antagonist candesartan blocks not only peripheral but also brain AT(1) receptors, prevents the hormonal and sympathoadrenal response to isolation stress and prevents the formation of stress-induced gastric ulcers. The mechanisms responsible for the prevention of stress-induced ulcers by the AT(1) receptor antagonist include protection from the stress-induced ischemia and inflammation (neutrophil infiltration and increase in ICAM-1 and TNF-alpha) in the gastric mucosa and a partial blockade of the stress-induced sympathoadrenal stimulation, while the protective effect of the glucocorticoid release during stress is maintained. AT(1) receptor antagonism prevents the stress-induced decrease in cortical CRH(1) and benzodiazepine binding and is anxiolytic. Blockade of brain angiotensin II AT(1) receptors offers a novel therapeutic opportunity for the treatment of anxiety and other stress-related disorders.
Collapse
Affiliation(s)
- Juan M Saavedra
- Section on Pharmacology, National Institute of Mental Health, National Institutes of Health, Department of Health and Human Services, Bethesda, MD 20892-1514, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Oliveira EM, Krieger JE. Chronic β-adrenoceptor stimulation and cardiac hypertrophy with no induction of circulating renin. Eur J Pharmacol 2005; 520:135-41. [PMID: 16129426 DOI: 10.1016/j.ejphar.2005.07.026] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2004] [Revised: 07/11/2005] [Accepted: 07/21/2005] [Indexed: 11/15/2022]
Abstract
The hemodynamic and cardiac effects of isoproterenol were examined in rats submitted to chronic salt loading (1% NaCl as drinking water) to prevent activation of the systemic renin-angiotensin system. Isoproterenol treatment for 1 week resulted in 36% and 44% (P<0.05) increase in left ventricle mass in both control and chronic salt loading rats and induction of cardiac angiotensin-converting enzyme activity and expression (P<0.05) with no changes in serum angiotensin-converting enzyme. Plasma renin activity decreased significantly with chronic salt loading and failed to increase by isoproterenol treatment, whereas it increased 2.33 fold (P<0.05) in animals kept on regular chow. Isoproterenol treatment leads to transient increase in heart rate and cardiac output while blood pressure remained unchanged. Altogether, these data provide evidence that isoproterenol induced hypertrophy is associated with cardiac induction of angiotensin-converting enzyme and daily transient hemodynamic overload even in the absence of systemic activation of renin-angiotensin system.
Collapse
Affiliation(s)
- Edilamar M Oliveira
- Laboratory of Biochemistry, School of Physical Education and Sports, University of São Paulo, São Paulo, SP, Brazil
| | | |
Collapse
|
50
|
Viatchenko-Karpinski S, Terentyev D, Jenkins LA, Lutherer LO, Györke S. Synergistic interactions between Ca2+ entries through L-type Ca2+ channels and Na+-Ca2+ exchanger in normal and failing rat heart. J Physiol 2005; 567:493-504. [PMID: 15975978 PMCID: PMC1474206 DOI: 10.1113/jphysiol.2005.091280] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
We used confocal Ca2+ imaging and the patch-clamp technique to investigate the interplay between Ca2+ entries through L-type Ca2+ channels (LCCs) and reverse-mode Na+-Ca2+ exchange (NCX) in activating Ca2+-induced Ca2+ release (CICR) from the sarcoplasmic reticulum (SR) in cardiac myocytes from normal and failing rat hearts. In normal myocytes exposed to N(6),2'-O-dibutyryl adenosine-3',5'-cyclic monophosphate (db-cAMP, membrane-permeable form of cAMP), the bell-shaped voltage dependence of cytosolic Ca2+ transients was dramatically broadened due to activation of SR Ca2+ release at high membrane potentials (30-120 mV). This broadening of Ca2+-transient voltage dependence could be prevented by KB-R7943, an inhibitor of the reverse-mode NCX. Trans-sarcolemmal Ca2+ entries were measured fluorometrically in myocytes during depolarizing steps to high membrane potentials. The total Ca2+ entry (deltaF(Tot)) was separated into two Ca2+ entry components, LCC-mediated (deltaF(LCC)) and NCX-mediated (deltaF(NCX)), by exposing the cells to the specific inhibitors of LCCs and reverse-mode NCX, nifedipine and KB-R7943, respectively. In the absence of protein kinase A (PKA) stimulation the amplitude of the Ca2+-inflow signal (deltaF(Tot)) corresponded to the arithmetic sum of the amplitudes of the KB-R7943- and nifedipine-resistant components (deltaF(Tot)=deltaF(LCC)+deltaF(NCX)). PKA activation resulted in significant increases in deltaF(Tot) and deltaF(LCC). Paradoxically, deltaF(Tot) became approximately threefold larger than the sum of the deltaF(NCX) and deltaF(LCC) components. In myocytes from failing hearts, stimulation of PKA failed to induce a shift in Ca2+ release voltage dependence toward more positive membrane potentials. Although the total and NCX-mediated Ca2+ entries were increased again, deltaF(Tot) did not significantly exceed the sum of deltaF(LCC) and deltaF(NCX). We conclude that the LCC and NCX Ca2+-entry pathways interact synergistically to trigger SR Ca2+ release on depolarization to positive membrane potentials in PKA-stimulated cardiac muscle. In heart failure, this new form of Ca2+ release is diminished and may potentially account for the compromised contractile performance and reduced functional reserve in failing hearts.
Collapse
Affiliation(s)
- Serge Viatchenko-Karpinski
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, Ohio State University Medical Center, 473 West 12th Avenue, Columbus, OH 43210-1252, USA
| | | | | | | | | |
Collapse
|