1
|
Fernandes-Costa F, Gomes da Silva RT, de Almeida AJPO, de Medeiros IA, de Assis Tafuri LS, Dos Santos GJ, Carlstrom M, Cruz JC. Organic vs. inorganic nitrates: Metabolic and vascular outcomes in STZ-induced diabetes in mice. Life Sci 2024; 359:123257. [PMID: 39561873 DOI: 10.1016/j.lfs.2024.123257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 11/09/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Diabetic animals often display dysregulated nitric oxide (NO) metabolism, contributing to vascular dysfunction. This study evaluates the metabolic and vascular effects of organic nitrate isosorbide mononitrate (ISMN) versus inorganic sodium nitrate (NaNO3) in mice with type 1 diabetes mellitus (T1DM) induced by streptozotocin (STZ). EXPERIMENTAL APPROACH T1DM was induced in male C57Bl6 mice with STZ ip and confirmed by fasting glucose. Mice were treated with ISMN (10 mg·kg-1) or NaNO3 (85 mg·L-1) for 14 days. A combination of in vivo, in vitro, and ex vivo studies assessed cardiometabolic benefits. RESULTS Both nitrates reduced blood and urinary hyperglycemia in T1DM mice, with ISMN exhibiting more significant reductions in blood glucose. ISMN and NaNO3 similarly reduced water and food intake, urinary volume, glucose intolerance, and insulin resistance while increasing insulin and nitrite levels in serum and urine. Both nitrates improved endothelium-independent vascular function and attenuated reactive oxygen species (ROS) while increasing NO levels in the aortic rings of T1DM mice. Furthermore, both nitrates similarly reduced mean arterial pressure in T1DM mice. CONCLUSION AND IMPLICATIONS ISMN and NaNO₃ have demonstrated comparable hypotensive and antioxidant effects, offering metabolic and vascular benefits in STZ-TDM1 mice. The more pronounced reduction in blood glucose with ISMN treatment compared to NaNO₃ is particularly promising. The antihyperglycemic effects of both nitrates were linked to increased serum insulin levels and enhanced insulin sensitivity. These results provide a foundation for future clinical studies to evaluate the potential of ISMN or NaNO3 as antidiabetogenic and antihypertensive adjuvant therapies in diabetic patients.
Collapse
Affiliation(s)
- Francineide Fernandes-Costa
- Graduate Program in Bioactive Synthetic and Natural Products, Center for Health Sciences, Federal University of Paraíba, João Pessoa, Brazil
| | | | | | - Isac Almeida de Medeiros
- Department of Pharmaceutical Sciences, Health Sciences Center, Federal University of Paraíba, João Pessoa, Brazil
| | | | - Gustavo Jorge Dos Santos
- Multicenter Graduate Program in Physiological Sciences, Department of Physiological Sciences, Center for Biological Sciences, Federal University of Santa Catarina - UFSC, Brazil
| | - Mattias Carlstrom
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
2
|
Fu Q, Wang Y, Yan C, Xiang YK. Phosphodiesterase in heart and vessels: from physiology to diseases. Physiol Rev 2024; 104:765-834. [PMID: 37971403 PMCID: PMC11281825 DOI: 10.1152/physrev.00015.2023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 10/17/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023] Open
Abstract
Phosphodiesterases (PDEs) are a superfamily of enzymes that hydrolyze cyclic nucleotides, including cyclic adenosine monophosphate (cAMP) and cyclic guanosine monophosphate (cGMP). Both cyclic nucleotides are critical secondary messengers in the neurohormonal regulation in the cardiovascular system. PDEs precisely control spatiotemporal subcellular distribution of cyclic nucleotides in a cell- and tissue-specific manner, playing critical roles in physiological responses to hormone stimulation in the heart and vessels. Dysregulation of PDEs has been linked to the development of several cardiovascular diseases, such as hypertension, aneurysm, atherosclerosis, arrhythmia, and heart failure. Targeting these enzymes has been proven effective in treating cardiovascular diseases and is an attractive and promising strategy for the development of new drugs. In this review, we discuss the current understanding of the complex regulation of PDE isoforms in cardiovascular function, highlighting the divergent and even opposing roles of PDE isoforms in different pathogenesis.
Collapse
Affiliation(s)
- Qin Fu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Key Laboratory for Drug Target Research and Pharmacodynamic Evaluation of Hubei Province, Wuhan, China
| | - Ying Wang
- Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chen Yan
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, New York, United States
| | - Yang K Xiang
- Department of Pharmacology, University of California at Davis, Davis, California, United States
- Department of Veterans Affairs Northern California Healthcare System, Mather, California, United States
| |
Collapse
|
3
|
Golzari-Sorkheh M, Liyanage I, Reed MA, Weaver DF. Alzheimer's Disease and COVID-19 Pathogenic Overlap: Implications for Drug Repurposing. Can J Neurol Sci 2024; 51:161-172. [PMID: 36991574 DOI: 10.1017/cjn.2023.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
As COVID-19 continues, a safe, cost-effective treatment strategy demands continued inquiry. Chronic neuroinflammatory disorders may appear to be of little relevance in this regard; often indolent and progressive disorders characterized by neuroinflammation (such as Alzheimer's disease (AD)) are fundamentally dissimilar in etiology and symptomology to COVID-19's rapid infectivity and pathology. However, the two disorders share extensive pathognomonic features, including at membrane, cytoplasmic, and extracellular levels, culminating in analogous immunogenic destruction of their respective organ parenchyma. We hypothesize that these mechanistic similarities may extent to therapeutic targets, namely that it is conceivable an agent against AD's immunopathy may have efficacy against COVID-19 and vice versa. It is notable that while extensively investigated, no agent has yet demonstrated significant therapeutic efficacy against AD's cognitive and memory declines. Yet this very failure has driven the development of numerous agents with strong mechanistic potential and clinical characteristics. Having already approved for clinical trials, these agents may be an expedient starting point in the urgent search for an effective COVID-19 therapy. Herein, we review the overlapping Alzheimer's/ COVID-19 targets and theorize several initial platforms.
Collapse
Affiliation(s)
| | - Imindu Liyanage
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Mark A Reed
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, ON, Canada
| | - Donald F Weaver
- Krembil Research Institute, University Health Network, Toronto, ON, Canada
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, ON, Canada
- Department of Chemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Glucosamine Use Is Associated with a Higher Risk of Cardiovascular Diseases in Patients with Osteoarthritis: Results from a Large Study in 685,778 Subjects. Nutrients 2022; 14:nu14183694. [PMID: 36145069 PMCID: PMC9506459 DOI: 10.3390/nu14183694] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 09/02/2022] [Accepted: 09/05/2022] [Indexed: 11/17/2022] Open
Abstract
Glucosamine is widely used around the world and as a popular dietary supplement and treatment in patients with osteoarthritis in China; however, the real-world cardiovascular risk of glucosamine in long-term use is still unclear. A retrospective, population-based cohort study was performed, based on the Beijing Medical Claim Data for Employees from 1 January 2010 to 31 December 2017. Patients newly diagnosed with osteoarthritis were selected and divided into glucosamine users and non- glucosamine users. The glucosamine users group was further divided into adherent, partially adherent, and non-adherent groups according to the medication adherence. New-onset cardiovascular diseases (CVD) events, coronary heart diseases (CHD), and stroke, were identified during the observational period. COX proportional regression models were used to estimate the risks. Of the 685,778 patients newly diagnosed with osteoarthritis including 240,419 glucosamine users and 445,359 non-users, the mean age was 56.49 (SD: 14.45) years and 59.35% were females. During a median follow-up of 6.13 years, 64,600 new-onset CVD, 26,530 CHD, and 17,832 stroke events occurred. Glucosamine usage was significantly associated with CVD (HR: 1.10; 95% CI: 1.08−1.11) and CHD (HR: 1.12; 95% CI: 1.09−1.15), but not with stroke (HR: 1.03; 95% CI: 0.99−1.06). The highest CVD risk was shown in the adherent group (HR: 1.68; 95% CI: 1.59−1.78), followed by the partially adherent group (HR: 1.26, 95% CI: 1.22−1.30), and the non-adherent group (HR: 1.03; 95% CI: 1.02−1.05), with a significant dose−response relationship (p-trend < 0.001). In this longitudinal study, adherent usage of glucosamine was significantly associated with a higher risk for cardiovascular diseases in patients with osteoarthritis.
Collapse
|
5
|
Cordes S, Mokhtari Z, Bartosova M, Mertlitz S, Riesner K, Shi Y, Mengwasser J, Kalupa M, McGeary A, Schleifenbaum J, Schrezenmeier J, Bullinger L, Diaz-Ricart M, Palomo M, Carrreras E, Beutel G, Schmitt CP, Beilhack A, Penack O. Endothelial damage and dysfunction in acute graft-versus-host disease. Haematologica 2021; 106:2147-2160. [PMID: 32675225 PMCID: PMC8327719 DOI: 10.3324/haematol.2020.253716] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
Clinical studies suggested that endothelial dysfunction and damage could be involved in the development and severity of acute graft-versus-host disease (aGVHD). Accordingly, we found increased percentage of apoptotic Casp3+ blood vessels in duodenal and colonic mucosa biopsies of patients with severe aGVHD. In murine experimental aGVHD, we detected severe microstructural endothelial damage and reduced endothelial pericyte coverage accompanied by reduced expression of endothelial tight junction proteins leading to increased endothelial leakage in aGVHD target organs. During intestinal aGVHD, colonic vasculature structurally changed, reflected by increased vessel branching and vessel diameter. Because recent data demonstrated an association of endothelium-related factors and steroid refractory aGVHD (SR-aGVHD), we analyzed human biopsies and murine tissues from SR-aGVHD. We found extensive tissue damage but low levels of alloreactive T cell infiltration in target organs, providing the rationale for T-cell independent SR-aGVHD treatment strategies. Consequently, we tested the endothelium-protective PDE5 inhibitor sildenafil, which reduced apoptosis and improved metabolic activity of endothelial cells in vitro. Accordingly, sildenafil treatment improved survival and reduced target organ damage during experimental SR-aGVHD. Our results demonstrate extensive damage, structural changes, and dysfunction of the vasculature during aGVHD. Therapeutic intervention by endothelium-protecting agents is an attractive approach for SR-aGVHD complementing current anti-inflammatory treatment options.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Shi
- Charité Universitätsmedizin Berlin
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Manrique-Acevedo C, Padilla J, Naz H, Woodford ML, Ghiarone T, Aroor AR, Hulse JL, Cabral-Amador FJ, Martinez-Diaz V, Hans CP, Whaley-Connell A, Martinez-Lemus LA, Lastra G. Mineralocorticoid Receptor in Myeloid Cells Mediates Angiotensin II-Induced Vascular Dysfunction in Female Mice. Front Physiol 2021; 12:588358. [PMID: 33854438 PMCID: PMC8039313 DOI: 10.3389/fphys.2021.588358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 02/17/2021] [Indexed: 11/13/2022] Open
Abstract
Enhanced mineralocorticoid receptor (MR) signaling is critical to the development of endothelial dysfunction and arterial stiffening. However, there is a lack of knowledge about the role of MR-induced adipose tissue inflammation in the genesis of vascular dysfunction in women. In this study, we hypothesize that MR activation in myeloid cells contributes to angiotensin II (Ang II)-induced aortic stiffening and endothelial dysfunction in females via increased pro-inflammatory (M1) macrophage polarization. Female mice lacking MR in myeloid cells (MyMRKO) were infused with Ang II (500 ng/kg/min) for 4 weeks. This was followed by determinations of aortic stiffness and vasomotor responses, as well as measurements of markers of inflammation and macrophage infiltration/polarization in different adipose tissue compartments. MyMRKO mice were protected against Ang II-induced aortic endothelial stiffening, as assessed via atomic force microscopy in aortic explants, and vasorelaxation dysfunction, as measured by aortic wire myography. In alignment, MyMRKO mice were protected against Ang II-induced macrophage infiltration and M1 polarization in visceral adipose tissue (VAT) and thoracic perivascular adipose tissue (tPVAT). Collectively, this study demonstrates a critical role of MR activation in myeloid cells in the pathogenesis of vascular dysfunction in females associated with pro-inflammatory macrophage polarization in VAT and tPVAT. Our data have potential clinical implications for the prevention and management of cardiovascular disease in women, who are disproportionally at higher risk for poor outcomes.
Collapse
Affiliation(s)
- Camila Manrique-Acevedo
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States
| | - Jaume Padilla
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Huma Naz
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States
| | - Makenzie L Woodford
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Nutrition and Exercise Physiology, University of Missouri, Columbia, MO, United States
| | - Thaysa Ghiarone
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Annayya R Aroor
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States
| | - Jack L Hulse
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States
| | | | - Vanesa Martinez-Diaz
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Chetan P Hans
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Division of Cardiovascular Medicine, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Adam Whaley-Connell
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States.,Division of Nephrology and Hypertension, Department of Medicine, University of Missouri, Columbia, MO, United States
| | - Luis A Martinez-Lemus
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, MO, United States.,Department of Biomedical, Biological and Chemical Engineering, University of Missouri, Columbia, MO, United States.,Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO, United States
| | - Guido Lastra
- Division of Endocrinology and Metabolism, Department of Medicine, University of Missouri, Columbia, MO, United States.,Research Service, Harry S. Truman Memorial Veterans' Hospital, Columbia, MO, United States
| |
Collapse
|
7
|
Bolea G, Philouze C, Dubois M, Risdon S, Humberclaude A, Ginies C, Charles AL, Geny B, Reboul C, Arnaud C, Dufour C, Meyer G. Digestive n-6 Lipid Oxidation, a Key Trigger of Vascular Dysfunction and Atherosclerosis in the Western Diet: Protective Effects of Apple Polyphenols. Mol Nutr Food Res 2021; 65:e2000487. [PMID: 33450108 DOI: 10.1002/mnfr.202000487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 01/04/2021] [Indexed: 01/07/2023]
Abstract
SCOPE A main risk factor of atherosclerosis is a Western diet (WD) rich in n-6 polyunsaturated fatty acids (PUFAs) sensitive to oxidation. Their oxidation can be initiated by heme iron of red meat leading to the formation of 4-hydroxy-2-nonenal (4-HNE), a cytotoxic aldehyde. An increased 4-HNE production is implicated in endothelial dysfunction and atherosclerosis. By contrast, a diet rich in proanthocyanidins reduces oxidative stress and arterial diseases. This study evaluates the effects of a WD on vascular integrity in ApolipoproteinE (ApoE-/- ) mice and the protective capacity of apple extract and puree rich in antioxidant proanthocyanidins. METHODS AND RESULTS ApoE-/- mice are fed during 12 weeks with a WD with or without n-6 PUFAs. Moreover, two WD + n-6 PUFAs groups are supplemented with apple puree or phenolic extract. An increase in digestive 4-HNE production associated with a rise in plasmatic 4-HNE and oxidized LDL concentrations is reported. Oxidizable n-6 PUFAs consumption is associated with a worsened endothelial dysfunction and atherosclerosis. Interestingly, supplementations with apple polyphenol extract or puree prevented these impairments while reducing oxidative stress. CONCLUSION n-6 lipid oxidation during digestion may be a key factor of vascular impairments. Nevertheless, an antioxidant strategy can limit 4-HNE formation during digestion and thus durably protect vascular function.
Collapse
Affiliation(s)
- Gaëtan Bolea
- EA4278 LaPEC, Laboratory of Cardiovascular Pharm-ecology, Avignon University, Avignon, F-84000, France
- INRAE, UMR408 SQPOV, Safety and Quality of Plant Products, Avignon University, Avignon, F-84000, France
| | - Clothilde Philouze
- EA4278 LaPEC, Laboratory of Cardiovascular Pharm-ecology, Avignon University, Avignon, F-84000, France
| | - Mathilde Dubois
- EA4278 LaPEC, Laboratory of Cardiovascular Pharm-ecology, Avignon University, Avignon, F-84000, France
| | - Sydney Risdon
- EA4278 LaPEC, Laboratory of Cardiovascular Pharm-ecology, Avignon University, Avignon, F-84000, France
| | - Anaïs Humberclaude
- EA4278 LaPEC, Laboratory of Cardiovascular Pharm-ecology, Avignon University, Avignon, F-84000, France
| | - Christian Ginies
- INRAE, UMR408 SQPOV, Safety and Quality of Plant Products, Avignon University, Avignon, F-84000, France
| | - Anne-Laure Charles
- UR3072, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072, Mitochondria, Oxidative Stress and Muscle Protection, University of Strasbourg, Strasbourg, F-67000, France
| | - Bernard Geny
- UR3072, Translational Medicine Federation of Strasbourg (FMTS), Faculty of Medicine, Team 3072, Mitochondria, Oxidative Stress and Muscle Protection, University of Strasbourg, Strasbourg, F-67000, France
| | - Cyril Reboul
- EA4278 LaPEC, Laboratory of Cardiovascular Pharm-ecology, Avignon University, Avignon, F-84000, France
| | - Claire Arnaud
- U1042 HP2, Cardiovascular and Respiratory Pathophysiology and Hypoxia, INSERM, Grenoble University, Grenoble, F-38000, France
| | - Claire Dufour
- INRAE, UMR408 SQPOV, Safety and Quality of Plant Products, Avignon University, Avignon, F-84000, France
| | - Grégory Meyer
- EA4278 LaPEC, Laboratory of Cardiovascular Pharm-ecology, Avignon University, Avignon, F-84000, France
| |
Collapse
|
8
|
Blanco-Rivero J, Xavier FE. Therapeutic Potential of Phosphodiesterase Inhibitors for Endothelial Dysfunction- Related Diseases. Curr Pharm Des 2021; 26:3633-3651. [PMID: 32242780 DOI: 10.2174/1381612826666200403172736] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 02/08/2020] [Indexed: 02/08/2023]
Abstract
Cardiovascular diseases (CVD) are considered a major health problem worldwide, being the main cause of mortality in developing and developed countries. Endothelial dysfunction, characterized by a decline in nitric oxide production and/or bioavailability, increased oxidative stress, decreased prostacyclin levels, and a reduction of endothelium-derived hyperpolarizing factor is considered an important prognostic indicator of various CVD. Changes in cyclic nucleotides production and/ or signalling, such as guanosine 3', 5'-monophosphate (cGMP) and adenosine 3', 5'-monophosphate (cAMP), also accompany many vascular disorders that course with altered endothelial function. Phosphodiesterases (PDE) are metallophosphohydrolases that catalyse cAMP and cGMP hydrolysis, thereby terminating the cyclic nucleotide-dependent signalling. The development of drugs that selectively block the activity of specific PDE families remains of great interest to the research, clinical and pharmaceutical industries. In the present review, we will discuss the effects of PDE inhibitors on CVD related to altered endothelial function, such as atherosclerosis, diabetes mellitus, arterial hypertension, stroke, aging and cirrhosis. Multiple evidences suggest that PDEs inhibition represents an attractive medical approach for the treatment of endothelial dysfunction-related diseases. Selective PDE inhibitors, especially PDE3 and PDE5 inhibitors are proposed to increase vascular NO levels by increasing antioxidant status or endothelial nitric oxide synthase expression and activation and to improve the morphological architecture of the endothelial surface. Thereby, selective PDE inhibitors can improve the endothelial function in various CVD, increasing the evidence that these drugs are potential treatment strategies for vascular dysfunction and reinforcing their potential role as an adjuvant in the pharmacotherapy of CVD.
Collapse
Affiliation(s)
- Javier Blanco-Rivero
- Departamento de Fisiologia, Facultad de Medicina, Universidad Autonoma de Madrid, Madrid, Spain
| | - Fabiano E Xavier
- Departamento de Fisiologia e Farmacologia, Centro de Biociencias, Universidade Federal de Pernambuco, Recife, Brazil
| |
Collapse
|
9
|
Zhao YX, Tong L, Zhang GM, Zhao XH, Sa YP, Liu Y, Lu DX, Ga Q, Wu P. L-Arginine Supplementation Improves Vascular Endothelial Dysfunction Induced by High-Fat Diet in Rats Exposed to Hypoxia. Wilderness Environ Med 2020; 31:400-406. [PMID: 33132032 DOI: 10.1016/j.wem.2020.06.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 06/01/2020] [Accepted: 06/17/2020] [Indexed: 10/23/2022]
Abstract
INTRODUCTION Our previous study showed that high-fat diet inhibited the increase in nitric oxide and endothelial nitric oxide synthase expression in the aortic endothelium of rats exposed to hypoxia, and hypoxia plus a high-fat diet led to earlier and more severe vascular endothelial dysfunction (VED) than hypoxia alone. The purpose of the present study was to investigate the effects of L-arginine on high-fat diet-induced VED of rats in hypoxia. METHODS Forty male Sprague-Dawley rats were randomly divided into 4 groups and treated with hypoxia (H group), hypoxia plus high-fat diet (H+HFD group), hypoxia plus L-arginine (H+L-Arg group), and hypoxia plus high-fat diet and L-arginine (H+HFD+L-Arg group) for 1 wk. Hypoxia was simulated in a hypobaric chamber with an altitude of 5000 m. Aortic morphology and endothelium-dependent vasorelaxation were used to assess VED. RESULTS High-fat diet impaired vascular remodeling and reduced endothelium-dependent vasodilator response to acetylcholine in rats exposed to hypoxia, secondary to dysregulation of the nitric oxide pathway. L-arginine supplementation significantly increased plasma nitrates and nitrites and endothelial nitric oxide synthase mRNA levels and improved ultrastructural changes in aortic endothelium and endothelium-dependent vasodilator response. CONCLUSIONS L-arginine prevents aortic ultrastructural changes and reverses VED induced by high-fat diet in rats exposed to hypoxia, which may have implications for VED induced by high-fat diet in high altitude dwellers.
Collapse
Affiliation(s)
- Yan-Xia Zhao
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China
| | - Li Tong
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China
| | - Guang-Mei Zhang
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China
| | - Xie-Hui Zhao
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China
| | - Yu-Ping Sa
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China
| | - Yan Liu
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China
| | - Dian-Xiang Lu
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China
| | - Qin Ga
- Research Center for High Altitude Medical Sciences, Qinghai University Medical College, Qinghai, China
| | - Ping Wu
- Department of Traditional Chinese Medicine, Qinghai University Medical College, Qinghai, China.
| |
Collapse
|
10
|
The Phosphodiesterase Type 5 Inhibitor Sildenafil Improves DNA Stability and Redox Homeostasis in Systemic Sclerosis Fibroblasts Exposed to Reactive Oxygen Species. Antioxidants (Basel) 2020; 9:antiox9090786. [PMID: 32854347 PMCID: PMC7555932 DOI: 10.3390/antiox9090786] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/11/2022] Open
Abstract
Systemic sclerosis (SSc) is a multi-system connective tissue disease characterized by the increased deposition of extracellular matrix proteins such as collagen and fibronectin. Although the pathogenesis is not completely understood, a number of studies suggest that free radicals could be the major contributors to the disease. Indeed, different studies demonstrated how oxidative stress could contribute to the fibrotic process activation at the level of the skin and visceral organs. Emerging evidences highlight the beneficial effects of sildenafil, a phosphodiesterase type 5 inhibitor (PDE5i), which protects different cell lines from the cell damage induced by reactive oxygen species (ROS). These data make sildenafil a good candidate for therapeutic treatment aimed to protect biological macromolecules against oxidative damage, thus preserving cell viability. The purpose of this study was to evaluate the sensitivity of SSc dermal fibroblasts to an oxidative insult and the ability for sildenafil to prevent/reduce the DNA damage due to ROS action. Additionally, we evaluated the capacity for sildenafil to influence redox homeostasis and cytotoxicity, as well as cell proliferation and cell cycle progression. We demonstrated that SSc fibroblasts have an increased sensitivity to a pro-oxidant environment in comparison to healthy controls. The sildenafil treatment reduced ROS-induced DNA damage, counteracted the negative effects of ROS on cell viability and proliferation, and promoted the activity of specific enzymes involved in redox homeostasis maintenance. To our knowledge, in this report, we demonstrate, for the first time, that sildenafil administration prevents ROS-induced instability in human dermal fibroblasts isolated by SSc patients. These results expand the use of PDE5i as therapeutic agents in SSc by indicating a protective role in tissue damage induced by oxidative insult.
Collapse
|
11
|
Abstract
Nitric oxide/cyclic guanosine monophosphate (cGMP) signaling is compromised in Alzheimer’s disease (AD), and phosphodiesterase 5 (PDE5), which degrades cGMP, is upregulated. Sildenafil inhibits PDE5 and increases cGMP levels. Integrating previous findings, we determine that most doses of sildenafil (especially low doses) likely activate peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α) via protein kinase G-mediated cyclic adenosine monophosphate (cAMP) response element binding protein (CREB) phosphorylation and/or Sirtuin-1 activation and PGC1α deacetylation. Via PGC1α signaling, low-dose sildenafil likely suppresses β-secretase 1 expression and amyloid-β (Aβ) generation, upregulates antioxidant enzymes, and induces mitochondrial biogenesis. Plus, sildenafil should increase brain perfusion, insulin sensitivity, long-term potentiation, and neurogenesis while suppressing neural apoptosis and inflammation. A systematic review of sildenafil in AD was undertaken. In vitro, sildenafil protected neural mitochondria from Aβ and advanced glycation end products. In transgenic AD mice, sildenafil was found to rescue deficits in CREB phosphorylation and memory, upregulate brain-derived neurotrophic factor, reduce reactive astrocytes and microglia, decrease interleukin-1β, interleukin-6, and tumor necrosis factor-α, decrease neural apoptosis, increase neurogenesis, and reduce tau hyperphosphorylation. All studies that tested Aβ levels reported significant improvements except the two that used the highest dosage, consistent with the dose-limiting effect of cGMP-induced phosphodiesterase 2 (PDE2) activation and cAMP depletion on PGC1α signaling. In AD patients, a single dose of sildenafil decreased spontaneous neural activity, increased cerebral blood flow, and increased the cerebral metabolic rate of oxygen. A randomized control trial of sildenafil (ideally with a PDE2 inhibitor) in AD patients is warranted.
Collapse
|
12
|
Negre-Salvayre A, Guerby P, Gayral S, Laffargue M, Salvayre R. Role of reactive oxygen species in atherosclerosis: Lessons from murine genetic models. Free Radic Biol Med 2020; 149:8-22. [PMID: 31669759 DOI: 10.1016/j.freeradbiomed.2019.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/16/2019] [Accepted: 10/16/2019] [Indexed: 12/19/2022]
Abstract
Atherosclerosis is a multifactorial chronic and inflammatory disease of medium and large arteries, and the major cause of cardiovascular morbidity and mortality worldwide. The pathogenesis of atherosclerosis involves a number of risk factors and complex events including hypercholesterolemia, endothelial dysfunction, increased permeability to low density lipoproteins (LDL) and their sequestration on extracellular matrix in the intima of lesion-prone areas. These events promote LDL modifications, particularly by oxidation, which generates acute and chronic inflammatory responses implicated in atherogenesis and lesion progression. Reactive oxygen species (ROS) (which include both free radical and non-free radical oxygen intermediates), play a key-role at each step of atherogenesis, in endothelial dysfunction, LDL oxidation, and inflammatory events involved in the initiation and development of atherosclerosis lesions. Most advanced knowledge supporting the "oxidative theory of atherosclerosis" i.e. the nature and the cellular sources of ROS and antioxidant defences, as well as the mechanisms involved in the redox balance, is based on the use of genetically engineered animals, i.e. transgenic, genetically modified, or altered for systems producing or neutralizing ROS in the vessels. This review summarizes the results obtained from animals genetically manipulated for various sources of ROS or antioxidant defences in the vascular wall, and their relevance (advance or limitation), for understanding the place and role of ROS in atherosclerosis.
Collapse
Affiliation(s)
| | - Paul Guerby
- Inserm U-1048, Université de Toulouse, France; Pôle de gynécologie obstétrique, Hôpital Paule-de-Viguier, CHU de Toulouse, France
| | | | | | | |
Collapse
|
13
|
Atalay HT, Ucgul AY, Turkcu UO, Ozmen MC, Yilmaz S, Bilgihan A. The Effect of Sildenafil on Selenite-Induced Cataract in Rats. Curr Eye Res 2020; 45:1082-1088. [PMID: 32023416 DOI: 10.1080/02713683.2020.1726405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE To investigate the effect of sildenafil on an experimental sodium selenite-induced cataract model in rats. MATERIALS AND METHODS Twenty-six young Wistar rats were separated into four groups. On postpartum day 10, six rats received only selenite (group 1, selenite-induced cataract), seven rats received selenite and high dose oral sildenafil (group 2, high-dose sildenafil-treated), seven rats received selenite and low dose oral sildenafil (group 3, low-dose sildenafil-treated), and six rats received only saline (group 4, controls). On postpartum day 30, cataract formation was graded and recorded using an operating microscope. The rats were sacrificed, lens tissues were isolated, and serum samples were collected. Nitrite oxide metabolites (NOx), advanced oxidative protein products (AOPP), and total sulfhydryl (TSH) levels were assessed in both serum and lenticular samples. RESULTS The rats treated with low-dose sildenafil showed lower levels of AOPP and NOx, and the higher levels of TSH than the rats in other experimental groups. Otherwise, the rats treated with high-dose sildenafil, similar to the selenite-induced cataract group, showed higher levels of AOPP and serum NOx than rats in the low-dose sildenafil-treated group. The rats treated with low-dose sildenafil also showed less cataract development than rats in the other experimental groups. CONCLUSION Low doses (0.7 mg/kg) of oral sildenafil might show a protective effect on cataract development by lowering oxidative stress.
Collapse
Affiliation(s)
- Hatice Tuba Atalay
- Department of Ophthalmology, Gazi University Medical School , Ankara, Turkey
| | - Ahmet Yucel Ucgul
- Department of Ophthalmology, Izzet Baysal Training and Research Hospital, Abant Izzet Baysal University , Bolu, Turkey
| | - Ummuhani Ozel Turkcu
- Department of Medical Biochemistry, Training and Research Hospital, Mugla Sıtkı Kocman University , Mugla, Turkey
| | - Mehmet Cuneyt Ozmen
- Department of Ophthalmology, Gazi University Medical School , Ankara, Turkey
| | - Samet Yilmaz
- Department of Medical Biochemistry, Gazi University Medical School , Ankara, Turkey
| | - Ayse Bilgihan
- Department of Medical Biochemistry, Gazi University Medical School , Ankara, Turkey
| |
Collapse
|
14
|
Friques AG, Santos FD, Angeli DB, Silva FAC, Dias AT, Aires R, Leal MA, Nogueira BV, Amorim FG, Campagnaro BP, Pereira TMC, Campos-Toimil M, Meyrelles SS, Vasquez EC. Bisphenol A contamination in infant rats: molecular, structural, and physiological cardiovascular changes and the protective role of kefir. J Nutr Biochem 2020; 75:108254. [DOI: 10.1016/j.jnutbio.2019.108254] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 08/15/2019] [Accepted: 09/16/2019] [Indexed: 12/11/2022]
|
15
|
Leal MAS, Aires R, Pandolfi T, Marques VB, Campagnaro BP, Pereira TMC, Meyrelles SS, Campos-Toimil M, Vasquez EC. Sildenafil reduces aortic endothelial dysfunction and structural damage in spontaneously hypertensive rats: Role of NO, NADPH and COX-1 pathways. Vascul Pharmacol 2019; 124:106601. [PMID: 31689530 DOI: 10.1016/j.vph.2019.106601] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 09/07/2019] [Accepted: 10/14/2019] [Indexed: 12/18/2022]
Abstract
Arterial hypertension is a condition associated with endothelial dysfunction, accompanied by an imbalance in the production of reactive oxygen species (ROS) and NO. The aim of this study was to investigate and elucidate the possible mechanisms of sildenafil, a selective phosphodiesterase-5 inhibitor, actions on endothelial function in aortas from spontaneously hypertensive rats (SHR). SHR treated with sildenafil (40 mg/kg/day, p.o., 3 weeks) were compared to untreated SHR and Wistar-Kyoto (WKY) rats. Systolic blood pressure (SBP) was measured by tail-cuff plethysmography and vascular reactivity was determined in isolated rat aortic rings. Circulating endothelial progenitor cells and systemic ROS were measured by flow cytometry. Plasmatic total antioxidant capacity, NO production and aorta lipid peroxidation were determined by spectrophotometry. Scanning electron microscopy was used for structural analysis of the endothelial surface. Sildenafil reduced high SBP and partially restored the vasodilator response to acetylcholine and sodium nitroprusside in SHR aortic rings. Using selective inhibitors, our experiments revealed an augmented participation of NO, with a simultaneous decrease of oxidative stress and of cyclooxygenase-1 (COX-1)-derived prostanoids contribution in the endothelium-dependent vasodilation in sildenafil-treated SHR compared to non-treated SHR. Also, the relaxant responses to sildenafil and 8-Br-cGMP were normalized in sildenafil-treated SHR and sildenafil restored the pro-oxidant/antioxidant balance and the endothelial architecture. In conclusion, sildenafil reverses endothelial dysfunction in SHR by improving vascular relaxation to acetylcholine with increased NO bioavailability, reducing the oxidative stress and COX-1 prostanoids, and improving cGMP/PKG signaling. Also, sildenafil reduces structural endothelial damage. Thus, sildenafil is a promising novel pharmacologic strategy to treat endothelial dysfunction in hypertensive states reinforcing its potential role as adjuvant in the pharmacotherapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Marcos A S Leal
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Rafaela Aires
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Thamirys Pandolfi
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Vinicius Bermond Marques
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Thiago M C Pereira
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil; Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil
| | - Silvana S Meyrelles
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Manuel Campos-Toimil
- Pharmacology of Chronic Diseases (CD PHARMA), Molecular Medicine and Chronic Diseases Research Centre (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain.
| | - Elisardo C Vasquez
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil; Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil
| |
Collapse
|
16
|
Naringin Reverses High-Cholesterol Diet-Induced Vascular Dysfunction and Oxidative Stress in Rats via Regulating LOX-1 and NADPH Oxidase Subunit Expression. BIOMED RESEARCH INTERNATIONAL 2019; 2019:3708497. [PMID: 31781614 PMCID: PMC6855071 DOI: 10.1155/2019/3708497] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/10/2019] [Indexed: 12/29/2022]
Abstract
Hypercholesterolaemia is associated with oxidative stress and endothelial dysfunction and leads to the development of atherosclerosis. Naringin exhibits cardiovascular protective and antioxidant properties. Therefore, the aim of this study was to assess the effect of naringin administration on vascular oxidative stress and endothelial dysfunction in hypercholesterolaemic rats and to elucidate its underlying mechanism. Sprague Dawley rats were fed a diet with 1.5% cholesterol (HCD) for 8 weeks to induce hypercholesterolaemia. Naringin (100 mg/kg body weight) was orally administrated to rats during the last 4 weeks of the diet treatment. After 8 weeks, the thoracic aorta was isolated to determine vascular function and nitric oxide (NO) levels. The aortic superoxide anion (O2−) level was detected using dihydroethidium (DHE) fluorescence staining. Protein expression of lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunits, and inducible nitric oxide synthase (iNOS), as well as oxidative damage markers, was also evaluated in aortae. Naringin treatment of hypercholesterolaemic rats enhanced aortic NO levels, restored endothelium-dependent responses to acetylcholine (ACh), and reduced aortic O2− levels. Furthermore, naringin treatment decreased LOX-1, NADPH oxidase subunits (p47phox, Nox2, and Nox4), and iNOS as well as oxidative damage markers (3-nitrotyrosine (3-NT) and 4-hydroxynonenal (4-HNE)) expression in aortic tissues from hypercholesterolaemic rats. These results demonstrate that naringin treatment improves endothelium dysfunction in hypercholesterolaemic rats, at least partially by decreasing oxidative stress via downregulation of LOX-1 and NADPH oxidase.
Collapse
|
17
|
Wortel RC, Mizrachi A, Li H, Markovsky E, Enyedi B, Jacobi J, Brodsky O, Cao J, Lippert AR, Incrocci L, Mulhall JP, Haimovitz-Friedman A. Sildenafil Protects Endothelial Cells From Radiation-Induced Oxidative Stress. J Sex Med 2019; 16:1721-1733. [PMID: 31585804 PMCID: PMC7269093 DOI: 10.1016/j.jsxm.2019.08.015] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 08/11/2019] [Accepted: 08/18/2019] [Indexed: 02/07/2023]
Abstract
Introduction: The etiology of radiation-induced erectile dysfunction (ED) is complex and multifactorial, and it appears to be mainly atherogenic. Aim: To focus on vascular aspects of radiation-induced ED and to elucidate whether the protective effects of sildenafil are mediated by attenuation of oxidative stress and apoptosis in the endothelial cells. Methods: Bovine aortic endothelial cells (BAECs), with or without pretreatment of sildenafil (5 μM at 5 minutes before radiation), were used to test endothelial dysfunction in response to external beam radiation at 10e15 Gy. Generation of reactive oxygen species (ROS) was studied. Extracellular hydrogen peroxide (H2O2) was measured using the Amplex Red assay and intracellular H2O2 using a fluorescent sensor. In addition, ROS superoxide (O2•-) was measured using a O2•- chemiluminescence enhancer. Both H2O2 and O2•- are known to reduce the bioavailability of nitric oxide, which is the most significant chemical mediator of penile erection. Generation of cellular peroxynitrite (ONOO−) was measured using a chemiluminescence assay with the PNCL probe. Subsequently, we measured the activation of acid sphingomyelinase (ASMase) enzyme by radioenzymatic assay using [14C-methylcholine] sphingomyelin as substrate, and the generation of the proapoptotic C16-ceramide was assessed using the diacylglycerol kinase assay. Endothelial cells apoptosis was measured as a readout of these cells’ dysfunction. Main Outcome Measures: Single high-dose radiation therapy induced NADPH oxidases (NOXs) activation and ROS generation via the proapoptotic ASMase/ceramide pathway. The radio-protective effect of sildenafil on BAECs was due to inhibition of this pathway. Results: Here, we demonstrate for the first time that radiation activated NOXs and induced generation of ROS in BAECs. In addition, we showed that sildenafil significantly reduced radiation-induced O2•- and as a result there was reduction in the generation of peroxynitrite in these cells. Subsequently, sildena fil protected the endothelial cells from radiation therapy-induced apoptosis. Strengths and Limitations: This is the first study demonstrating that single high-dose radiation therapy induced NOXs activation, resulting in the generation of O2•- and peroxynitrite in endothelial cells. Sildenafil reduced ROS generation by inhibiting the ASMase/ceramide pathway. These studies should be followed in an animal model of ED. Conclusions: This study demonstrated that sildenafil protects BAECs from radiation-induced oxidative stress by reducing NOX-induced ROS generation, thus resulting in decreased endothelial dysfunction. Therefore, it provides a potential mechanism to better understand the atherogenic etiology of postradiation ED.
Collapse
Affiliation(s)
- R C Wortel
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Radiation Oncology, Department of Urology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - A Mizrachi
- Head and Neck Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Otolaryngology Head and Neck Surgery, Rabin Medical Center, and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - H Li
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Markovsky
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - B Enyedi
- Department of Cell Biology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Physiology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - J Jacobi
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Hematopathology, Columbia University Medical Center, New York, NY, USA
| | - O Brodsky
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Urology, Hadassah Medical Center, Jerusalem, Israel
| | - J Cao
- Department of Chemistry, Southern Methodist University, Dallas, TX, USA
| | - A R Lippert
- Department of Chemistry, Southern Methodist University, Dallas, TX, USA
| | - L Incrocci
- Department of Radiation Oncology, Erasmus MC, Rotterdam, The Netherlands
| | - J P Mulhall
- Department of Urology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Haimovitz-Friedman
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
18
|
Oboh G, Adebayo AA, Ademosun AO. HPLC phenolic fingerprinting, antioxidant and anti-phosphodiesterase-5 properties of Rauwolfia vomitoria extract. J Basic Clin Physiol Pharmacol 2019; 30:jbcpp-2019-0059. [PMID: 31469654 DOI: 10.1515/jbcpp-2019-0059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 07/27/2019] [Indexed: 06/10/2023]
Abstract
Background In Nigerian traditional medicine, Rauwolfia vomitoria has been reported to be useful in the management of various human diseases, but there is no relevant information to substantiate its involvement in managing diseases arising from vascular dysfunction and oxidative stress. However, this study sought to investigate the antioxidant property of R. vomitoria and its effect on phophodiesterase-5 activity in vitro. Methods The antioxidant property was assessed through ferric-reducing antioxidant power (FRAP), copper chelation, and ABTS radical-scavenging activity. In addition, the effect of R. vomitoria on phosphodiesterase-5 (PDE-5) activity was assessed in vitro. Furthermore, analysis of phenolic compounds present in R. vomitoria was carried out using high-performance liquid chromatography (HPLC). Results The findings in this study revealed that R. vomitoria inhibited PDE-5 in a dose-dependent manner (IC50 = 252.42 μg/mL). Furthermore, the antioxidant activity of R. vomitoria was established through FRAP (19.68 mg AAE/g), ABTS radical-scavenging ability (74.25 mmol TEAC/g), and Cu2+-chelating ability (IC50 = 0.13 mg/mL). Conclusions The antioxidant property of R. vomitoria and its inhibitory effect on PDE-5 could be useful in the management of diseases arising from vascular dysfunction and oxidative stress.
Collapse
Affiliation(s)
- Ganiyu Oboh
- Functional Foods and Nutraceutical Research Unit, Biochemistry Department, Federal University of Technology, Akure 340001, Nigeria
| | - Adeniyi A Adebayo
- Functional Foods and Nutraceutical Research Unit, Biochemistry Department, Federal University of Technology, Akure, Nigeria
| | - Ayokunle O Ademosun
- Functional Foods and Nutraceutical Research Unit, Biochemistry Department, Federal University of Technology, Akure, Nigeria
| |
Collapse
|
19
|
Chisté LA, Pereira BP, Porto ML, de Oliveira JP, de Assis ALEM, Nogueira BV, Meyrelles SS, de Andrade TU, Campos-Toimil M, Vasquez EC, Campagnaro BP, Pereira TMC. Worsening of Oxidative Stress, DNA Damage, and Atherosclerotic Lesions in Aged LDLr -/- Mice after Consumption of Guarana Soft Drinks. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9042526. [PMID: 31281596 PMCID: PMC6590538 DOI: 10.1155/2019/9042526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/12/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND Excessive consumption of soft drinks (SD) has become a health problem worldwide due to its association with related cardiovascular diseases. We investigated the possible impacts associated with the consumption of Brazilian guarana (normal and zero) SD in dyslipidemic mice, thus mitigating potential clinical confounders such as poor-quality diet, lifestyle, body composition, and/or comorbidities. METHODS Sixteen-month-old LDLr-/- mice were divided into the following groups: (1) control; (2) GSD: normal guarana SD; and (3) Z-GSD: zero guarana SD. All were fed ad libitum, and blood pressure was measured noninvasively. After 8 weeks, aorta, blood, liver, and stomach samples were collected for histological and biochemical analyses. RESULTS Guarana soft drinks increased atherosclerosis (~60%) and were associated with hypercholesterolemia, hypertension, oxidative stress, DNA fragmentation, and apoptosis (~2-fold) of blood cells, besides presenting an increase in liver and gastric damage even in normoglycemia. Interestingly, Z-GSD did not cause the aforementioned changes, except in hemodynamic and renal parameters. CONCLUSIONS Chronic administration of GSD is prooxidative, compromising the cardiovascular, gastric, and hepatic systems; the effects are due at least in part to free sugar consumption but not to guarana extract per se.
Collapse
Affiliation(s)
- Layla Aparecida Chisté
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil
| | - Beatriz Peters Pereira
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil
| | - Marcella Leite Porto
- Federal Institute of Education, Science, and Technology (IFES), Vila Velha, ES, Brazil
| | - Jairo Pinto de Oliveira
- Laboratory of Cellular Ultrastructure Carlos Alberto Redins (LUCCAR), Department of Morphology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, ES, Brazil
| | - Arícia Leone Evangelista Monteiro de Assis
- Laboratory of Cellular Ultrastructure Carlos Alberto Redins (LUCCAR), Department of Morphology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, ES, Brazil
| | - Breno Valentim Nogueira
- Laboratory of Cellular Ultrastructure Carlos Alberto Redins (LUCCAR), Department of Morphology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, ES, Brazil
| | - Silvana Santos Meyrelles
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | - Tadeu Uggere de Andrade
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil
| | - Manuel Campos-Toimil
- Pharmacology of Chronic Diseases (CDPHARMA), Molecular Medicine and Chronic Diseases Research Center (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Elisardo Corral Vasquez
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil
| | | | - Thiago Melo Costa Pereira
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil
- Federal Institute of Education, Science, and Technology (IFES), Vila Velha, ES, Brazil
- Pharmacology of Chronic Diseases (CDPHARMA), Molecular Medicine and Chronic Diseases Research Center (CIMUS), University of Santiago de Compostela, Santiago de Compostela, Spain
| |
Collapse
|
20
|
Teixeira-da-Silva JJ, Nunes-Moreira HS, Silva CO, Lahlou S, Naro F, Xavier FE, Duarte GP. Chronic administration of sildenafil improves endothelial function in spontaneously hypertensive rats by decreasing COX-2 expression and oxidative stress. Life Sci 2019; 225:29-38. [DOI: 10.1016/j.lfs.2019.03.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Revised: 03/20/2019] [Accepted: 03/29/2019] [Indexed: 11/28/2022]
|
21
|
Antioxidant effect of sildenafil: Potential hepatoprotection via differential expression of mitochondrial proteins in apolipoprotein E knockout mice. Pharmacol Rep 2019; 71:422-429. [PMID: 31003152 DOI: 10.1016/j.pharep.2019.01.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 12/12/2018] [Accepted: 01/04/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND High plasma cholesterol levels are able to trigger several pathophysiological events, including inflammation, cell damage and especially oxidative stress. Previously, studies have shown that sildenafil exhibited antioxidant effects in several experimental models. Here we evaluate the role of sildenafil in liver redox equilibrium of apolipoprotein E knockout (apoE-KO) mice. METHODS ApoE-KO mice were divided in two groups: one group received the PDE5 inhibitor sildenafil (40 mg/kg/day) for 3 weeks (apoE-KO + Sil) and was compared to a second group of apoE-KO mice, which received only the vehicle (water) for 3 weeks (apoE-KO). Control group (C57 mice) received only a standard chow diet. At the age of 18 weeks, mice livers were collected for the measurement of intracellular ROS levels and apoptotic cells by flow cytometry analysis, and mitochondria isolation for proteomic analysis. RESULTS Compared to the control group, liver cells from apoE-KO presented some typical redox imbalance features: higher levels of intracellular ROS (global oxidative stress ˜60%, superoxide anion ˜82%, and peroxynitrite/hydroxyl radical ˜53%), higher amounts of apoptotic cells (up to ˜19%) and higher mitochondrial intensity of catalase (+339%) and transferrin spots (+914%). After treatment with sildenafil, apoE-KO presented ROS levels and the number of apoptotic cells similar to those observed in C57. In addition, when compared to apoE-KO, apoE-KO + Sil showed lower spots volumes of catalase (-23%) and transferrin (-71%) and up-regulation of urate oxidase (+94%). CONCLUSION The treatment with sildenafil is able to induce beneficial changes in liver mitochondrial protein dynamics, which restores the redox homeostasis contributing to a potential hepatoprotection.
Collapse
|
22
|
Oboh G, Adebayo AA, Ademosun AO. Hunteria umbellata seed extract administration modulates activities of phosphodiesterase-5 and purinergic enzymes relevant to erection in normal male rats. ACTA ACUST UNITED AC 2019. [DOI: 10.1007/s13596-019-00368-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
23
|
Jacinto TA, Meireles GS, Dias AT, Aires R, Porto ML, Gava AL, Vasquez EC, Pereira TMC, Campagnaro BP, Meyrelles SS. Increased ROS production and DNA damage in monocytes are biomarkers of aging and atherosclerosis. Biol Res 2018; 51:33. [PMID: 30185234 PMCID: PMC6123971 DOI: 10.1186/s40659-018-0182-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 08/31/2018] [Indexed: 01/11/2023] Open
Abstract
Background New evidence demonstrates that aging and dyslipidemia are closely associated with oxidative stress, DNA damage and apoptosis in some cells and extravascular tissues. However, in monocytes, which are naturally involved in progression and/or resolution of plaque in atherosclerosis, this concurrence has not yet been fully investigated. In this study, we evaluated the influence of aging and hypercholesterolemia on serum pro-inflammatory cytokines, oxidative stress, DNA damage and apoptosis in monocytes from apolipoprotein E-deficient (apoE−/−) mice compared with age-matched wild-type C57BL/6 (WT) mice. Experiments were performed in young (2-months) and in old (18-months) male wild-type (WT) and apoE−/− mice. Results Besides the expected differences in serum lipid profile and plaque formation, we observed that atherosclerotic mice exhibited a significant increase in monocytosis and in serum levels of pro-inflammatory cytokines compared to WT mice. Moreover, it was observed that the overproduction of ROS, led to an increased DNA fragmentation and, consequently, apoptosis in monocytes from normocholesterolemic old mice, which was aggravated in age-matched atherosclerotic mice. Conclusions In this study, we demonstrate that a pro-inflammatory systemic status is associated with an impairment of functionality of monocytes during aging and that these parameters are fundamental extra-arterial contributors to the aggravation of atherosclerosis. The present data open new avenues for the development of future strategies with the purpose of treating atherosclerosis.
Collapse
Affiliation(s)
- Thais A Jacinto
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
| | - Giselle S Meireles
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Rua Mercúrio, s/n, Boa Vista 1, Vila Velha, ES, 29102-623, Brazil
| | - Ananda T Dias
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
| | - Rafaela Aires
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
| | - Marcella L Porto
- Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil
| | - Agata L Gava
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil.,Division of Nephrology, McMaster University, Hamilton, ON, Canada
| | - Elisardo C Vasquez
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil.,Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Rua Mercúrio, s/n, Boa Vista 1, Vila Velha, ES, 29102-623, Brazil
| | - Thiago Melo C Pereira
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Rua Mercúrio, s/n, Boa Vista 1, Vila Velha, ES, 29102-623, Brazil.,Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil
| | - Bianca P Campagnaro
- Laboratory of Translational Physiology and Pharmacology, Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Rua Mercúrio, s/n, Boa Vista 1, Vila Velha, ES, 29102-623, Brazil.
| | - Silvana S Meyrelles
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
| |
Collapse
|
24
|
Modulatory effect of some citrus (Citrus limon, Citrus reticulata, Citrus maxima) peels on monoamine oxidase, phosphodiesterase-5 and angiotensin-1 converting enzyme activities in rat heart homogenate. ACTA ACUST UNITED AC 2018; 16:jcim-2018-0067. [DOI: 10.1515/jcim-2018-0067] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023]
Abstract
Abstract
Background
Citrus peels have been reported useful in folk medicine for the management of cardiovascular diseases, but there is dearth of information on the possible mechanisms for their therapeutic action. The aim of this study was to investigate the effect of methanolic extracts from some citrus [lime (Citrus limon), tangerine (Citrus reticulata), shaddock (Citrus maxima)] peels on some enzymes relevant to the management of cardiovascular diseases [monoamine oxidase (MAO), phosphodiesterase-5 (PDE-5) and angiotensin-1-converting enzyme (ACE)].
Methods
Effect of methanolic extracts of lime, tangerine and shaddock peels on MAO, PDE-5 and ACE were carried out using standard methods. In addition, the ability of the extracts to prevent oxidative damage in rat heart homogenates was also investigated. Finally, the total polyphenol content of extracts was determined.
Results
The results revealed that methanolic extracts of lime, tangerine and shaddock peels inhibited MAO, PDE-5, ACE and pro-oxidants induced lipid peroxidation in rat heart homogenate in a concentration-dependent manner.
Conclusions
Findings in this study revealed citrus peel methanolic extracts as natural inhibitor of enzymes (MAO, PDE-5 and ACE) implicated in cardiovascular diseases. Therefore, citrus peels could help in the management of cardiovascular diseases possibly through inhibition of these enzymes.
Collapse
|
25
|
Wobst J, Schunkert H, Kessler T. Genetic alterations in the NO-cGMP pathway and cardiovascular risk. Nitric Oxide 2018; 76:105-112. [PMID: 29601927 DOI: 10.1016/j.niox.2018.03.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 03/18/2018] [Accepted: 03/26/2018] [Indexed: 12/18/2022]
Abstract
In the past ten years, several chromosomal loci have been identified by genome-wide association studies to influence the risk of coronary artery disease (CAD) and its risk factors. The GUCY1A3 gene encoding the α1 subunit of the soluble guanylyl cyclase (sGC) resides at one of these loci and has been strongly associated with blood pressure and CAD risk. More recently, further genes in the pathway encoding the endothelial nitric oxide synthase, the phosphodiesterases 3A and 5A, and the inositol 1,4,5-trisphosphate receptor I-associated protein (IRAG), i.e., NOS3, PDE3A, PDE5A, and MRVI1, respectively, were likewise identified as CAD risk genes. In this review, we highlight the genetic findings linking variants in NO-cGMP signaling and cardiovascular disease, discuss the potential underlying mechanisms which might propagate the development of atherosclerosis, and speculate about therapeutic implications.
Collapse
Affiliation(s)
- Jana Wobst
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany
| | - Heribert Schunkert
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany
| | - Thorsten Kessler
- Deutsches Herzzentrum München, Klinik für Herz- und Kreislauferkrankungen, Technische Universität München, Munich, Germany; Deutsches Zentrum für Herz-Kreislauf-Forschung (DZHK) e.V., partner site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
26
|
Molehin OR, Adeyanju AA, Adefegha SA, Aina OO, Afolabi BA, Olowoyeye AO, Oyediran JA, Oladiran OR. Sildenafil, a phosphodiesterase-5 inhibitor, offers protection against carbon tetrachloride-induced hepatotoxicity in rat. J Basic Clin Physiol Pharmacol 2018; 29:29-35. [PMID: 29283882 DOI: 10.1515/jbcpp-2017-0011] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 10/25/2017] [Indexed: 05/20/2023]
Abstract
BACKGROUND Elevation of phosphodiesterase-5 (PDE5) activity converts cyclic guanosine monophosphate (cGMP) to 5'-GMP, a mechanism that could be associated with drug-mediated hepatotoxicity. This study investigated whether selective inhibition of PDE5 by sildenafil could offer protection against hepatotoxicity induced by carbon tetrachloride (CCl4). METHODS CCl4 (0.5 mL/kg) was administered intraperitoneally to induce hepatotoxicity. The control group received normal saline. Sildenafil (5 mg, 10 mg, and 20 mg/kg, p.o.) was administered to CCl4-treated rats. RESULTS CCl4 significantly increased the serum levels of gamma glutamyl transferase (γ-GT), alkaline phosphatase (ALP), aspartate aminotransferase (AST), and alanine aminotransferase (ALT) and reduced total protein (TP) (p<0.05). Pretreatment with sildenafil moderately reduced ALP, AST, and ALT activities with modest increase in TP level. CCl4-induced changes in the antioxidant status of the liver were significantly improved by sildenafil, especially at the lowest dose of 5 mg/kg by elevating the levels of reduced glutathione (GSH), glutathione peroxidase (GPx), catalase (CAT), superoxide dismutase (SOD), and glutathione-S-transferase (GST) and preventing lipid peroxidation (p<0.05). Sildenafil did not significantly alter the total cholesterol and triglyceride levels. However, high-density lipoprotein (HDL) level was significantly increased by sildenafil (p<0.05). CONCLUSIONS The results from this study suggest that sildenafil, when used at low doses, may be a useful pharmacological protective agent against CCl4-induced hepatotoxicity.
Collapse
Affiliation(s)
- Olorunfemi R Molehin
- Department of Biochemistry, Faculty of Science, Ekiti State University, Ado-Ekiti. P.M.B.5363, Ado-Ekiti, Nigeria, Phone: +234 803 462 1267, E-mail:
| | - Anne A Adeyanju
- Department of Biological Sciences, McPherson University, Seriki Sotayo, Ajebo, Nigeria
| | - Stephen A Adefegha
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology, Akure, Nigeria
| | - Oluwasanmi O Aina
- Department of Veterinary Anatomy, University of Ibadan, Ibadan, Nigeria
| | | | - Ayorinde O Olowoyeye
- Department of Biochemistry, Faculty of Science, Ekiti State University, Ado-Ekiti. P.M.B.5363, Ado-Ekiti, Nigeria
| | - Jesutomi A Oyediran
- Department of Biochemistry, Faculty of Science, Ekiti State University, Ado-Ekiti. P.M.B.5363, Ado-Ekiti, Nigeria
| | - Opeyemi R Oladiran
- Department of Biochemistry, Faculty of Science, Ekiti State University, Ado-Ekiti. P.M.B.5363, Ado-Ekiti, Nigeria
| |
Collapse
|
27
|
Luo S, Yang M, Jin H, Xu ZQ, Li YF, Xia P, Yang YR, Chen BC, Zhang Y. The role of sildenafil in the development of transplant arteriosclerosis in rat aortic grafts. Am J Transl Res 2017; 9:4914-4924. [PMID: 29218089 PMCID: PMC5714775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 05/01/2017] [Indexed: 06/07/2023]
Abstract
Chronic rejection (CR), which is characterized histologically by progressive graft arteriosclerosis, remains a significant barrier to the long-term survival of a graft. Sildenafil has been shown to protect vascular endothelial cells. In this study, we found that sildenafil significantly reduces the thickness of transplant vascular intima in a rat aortic transplant model. Moreover, sildenafil dramatically decreased the expression of transforming growth factor-β1 (TGF-β1), vascular endothelial growth factor (VEGF), and α-smooth muscle actin (α-SMA) in the grafted aortas and increased the concentrations of cyclic guanosine monophosphate (cGMP) and endothelial nitric oxide synthase (eNOS) in serum. Furthermore, the ratio of regulatory T (Treg) cells and the expression of FoxP3 were increased, and the ratio of Th17 cells was decreased in the sildenafil-treated group. These results demonstrate that sildenafil enhances nitric oxide (NO) signaling by increasing the availability of cGMP, leading to an increase in the ratio of Treg/Th17 cells to attenuate transplant arteriosclerosis in a rat aortic transplant model.
Collapse
Affiliation(s)
- Shuai Luo
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
- Department of Urology, Huangshi Central HospitalHuangshi 435000, Hubei Province, China
| | - Mei Yang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Hao Jin
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Zi-Qiang Xu
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Yi-Fu Li
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Peng Xia
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Yi-Rrong Yang
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Bi-Cheng Chen
- Zhejiang Provincial Top Key Discipline in Surgery, Wenzhou Key Laboratory of Surgery, Department of Surgery, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| | - Yan Zhang
- Transplantation Centre, The First Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325015, Zhejiang Province, China
| |
Collapse
|
28
|
Coutinho PN, Pereira BP, Hertel Pereira AC, Porto ML, Monteiro de Assis ALE, Côgo Destefani A, Meyrelles SS, Vasquez EC, Nogueira BV, de Andrade TU, Endringer DC, Fronza M, Costa Pereira TM. Chronic administration of antioxidant resin from Virola oleifera attenuates atherogenesis in LDLr -/- mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 206:65-72. [PMID: 28502908 DOI: 10.1016/j.jep.2017.05.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 04/25/2017] [Accepted: 05/10/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Virola oleifera (Schott) A. C. Smith, Myristicaceae has been largely used in traditional folk medicine in Brazil as an anti-inflammatory agent and our previous data indicated the antioxidant properties in other oxidative stress-related models. However, its effects on atherosclerosis (AT) are not yet investigated. AIMS OF THE STUDY To evaluate the influence of resin from Virola oleifera (RV) on progression of AT in LDLr-/- mice. MATERIALS AND METHODS LDLr-/- mice were divided into 4 groups: 1) The ND group received a normal diet without treatment. 2) The HD group received a high-fat diet without treatment. 3) The HD-V50 received a high-fat diet and was orally treated with RV at 50mg/Kg. 4) The HD-V300 received a high-fat diet and was orally treated with RV at 300mg/Kg. After 4 weeks, blood was collected to quantify biochemical parameters and ROS total and the aorta was removed to measure the lipid deposition by en face analysis. The liver was also collected to determine total lipids and lipid and protein oxidation. In order to investigate in more detail the contributions of RV in the vascular structure, we carried out the in vitro tests using four cellular types: macrophages, fibroblasts, vascular smooth muscle and endothelial cells. RESULTS We showed that the chronic treatment of RV at both doses reduced vascular lipid accumulation (~50%, p<0.05), probably through systemic and hepatic antioxidant effects, independent of dyslipidemia. Moreover, the in vitro assay results demonstrated that RV develops antioxidant properties on the vascular smooth muscle and endothelial cells, reinforcing the protective role of RV in progression of AT. LPS-stimulated macrophages treated with RV resulted in a significant reduction of NO production in a concentration-dependent manner. CONCLUSIONS Chronic treatment with RV diminishes lipid deposition in atherosclerotic mice, which may be justified, at least in part, by antioxidant systemic and local mechanisms, reinforcing the protective role this resin in the setting of vascular lipid deposition, independent of hypercholesterolemia.
Collapse
Affiliation(s)
- Paola Nogueira Coutinho
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil.
| | - Beatriz Peters Pereira
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil.
| | | | - Marcella Leite Porto
- Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil.
| | - Arícia Leone Evangelista Monteiro de Assis
- Laboratory of Cellular Ultrastructure Carlos Alberto Redins (LUCCAR), Department of Morphology, UFES, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Afrânio Côgo Destefani
- Laboratory of Cellular Ultrastructure Carlos Alberto Redins (LUCCAR), Department of Morphology, UFES, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Silvana Santos Meyrelles
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Elisardo Corral Vasquez
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil; Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Breno Valentim Nogueira
- Laboratory of Cellular Ultrastructure Carlos Alberto Redins (LUCCAR), Department of Morphology, UFES, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Tadeu Uggere de Andrade
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil.
| | - Denise Coutinho Endringer
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil; Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil.
| | - Marcio Fronza
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil.
| | - Thiago Melo Costa Pereira
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil; Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil.
| |
Collapse
|
29
|
Sfyri P, Matsakas A. Crossroads between peripheral atherosclerosis, western-type diet and skeletal muscle pathophysiology: emphasis on apolipoprotein E deficiency and peripheral arterial disease. J Biomed Sci 2017; 24:42. [PMID: 28688452 PMCID: PMC5502081 DOI: 10.1186/s12929-017-0346-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 06/07/2017] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory process that, in the presence of hyperlipidaemia, promotes the formation of atheromatous plaques in large vessels of the cardiovascular system. It also affects peripheral arteries with major implications for a number of other non-vascular tissues such as the skeletal muscle, the liver and the kidney. The aim of this review is to critically discuss and assimilate current knowledge on the impact of peripheral atherosclerosis and its implications on skeletal muscle homeostasis. Accumulating data suggests that manifestations of peripheral atherosclerosis in skeletal muscle originates in a combination of increased i)-oxidative stress, ii)-inflammation, iii)-mitochondrial deficits, iv)-altered myofibre morphology and fibrosis, v)-chronic ischemia followed by impaired oxygen supply, vi)-reduced capillary density, vii)- proteolysis and viii)-apoptosis. These structural, biochemical and pathophysiological alterations impact on skeletal muscle metabolic and physiologic homeostasis and its capacity to generate force, which further affects the individual's quality of life. Particular emphasis is given on two major areas representing basic and applied science respectively: a)-the abundant evidence from a well-recognised atherogenic model; the Apolipoprotein E deficient mouse and the role of a western-type diet and b)-on skeletal myopathy and oxidative stress-induced myofibre damage from human studies on peripheral arterial disease. A significant source of reactive oxygen species production and oxidative stress in cardiovascular disease is the family of NADPH oxidases that contribute to several pathologies. Finally, strategies targeting NADPH oxidases in skeletal muscle in an attempt to attenuate cellular oxidative stress are highlighted, providing a better understanding of the crossroads between peripheral atherosclerosis and skeletal muscle pathophysiology.
Collapse
Affiliation(s)
- Peggy Sfyri
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom
| | - Antonios Matsakas
- Molecular Physiology Laboratory, Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, University of Hull, Cottingham Road, Hull, HU6 7RX, United Kingdom.
| |
Collapse
|
30
|
The Therapeutic Potential of Anti-Inflammatory Exerkines in the Treatment of Atherosclerosis. Int J Mol Sci 2017; 18:ijms18061260. [PMID: 28608819 PMCID: PMC5486082 DOI: 10.3390/ijms18061260] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/22/2017] [Accepted: 06/09/2017] [Indexed: 12/15/2022] Open
Abstract
Although many cardiovascular (CVD) medications, such as antithrombotics, statins, and antihypertensives, have been identified to treat atherosclerosis, at most, many of these therapeutic agents only delay its progression. A growing body of evidence suggests physical exercise could be implemented as a non-pharmacologic treatment due to its pro-metabolic, multisystemic, and anti-inflammatory benefits. Specifically, it has been discovered that certain anti-inflammatory peptides, metabolites, and RNA species (collectively termed “exerkines”) are released in response to exercise that could facilitate these benefits and could serve as potential therapeutic targets for atherosclerosis. However, much of the relationship between exercise and these exerkines remains unanswered, and there are several challenges in the discovery and validation of these exerkines. This review primarily highlights major anti-inflammatory exerkines that could serve as potential therapeutic targets for atherosclerosis. To provide some context and comparison for the therapeutic potential of exerkines, the anti-inflammatory, multisystemic benefits of exercise, the basic mechanisms of atherosclerosis, and the limited efficacies of current anti-inflammatory therapeutics for atherosclerosis are briefly summarized. Finally, key challenges and future directions for exploiting these exerkines in the treatment of atherosclerosis are discussed.
Collapse
|
31
|
Xu J, Mukerjee S, Silva-Alves CRA, Carvalho-Galvão A, Cruz JC, Balarini CM, Braga VA, Lazartigues E, França-Silva MS. A Disintegrin and Metalloprotease 17 in the Cardiovascular and Central Nervous Systems. Front Physiol 2016; 7:469. [PMID: 27803674 PMCID: PMC5067531 DOI: 10.3389/fphys.2016.00469] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 09/30/2016] [Indexed: 01/19/2023] Open
Abstract
ADAM17 is a metalloprotease and disintegrin that lodges in the plasmatic membrane of several cell types and is able to cleave a wide variety of cell surface proteins. It is somatically expressed in mammalian organisms and its proteolytic action influences several physiological and pathological processes. This review focuses on the structure of ADAM17, its signaling in the cardiovascular system and its participation in certain disorders involving the heart, blood vessels, and neural regulation of autonomic and cardiovascular modulation.
Collapse
Affiliation(s)
- Jiaxi Xu
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | - Snigdha Mukerjee
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | | | | | - Josiane C Cruz
- Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Camille M Balarini
- Centro de Ciências da Saúde, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Valdir A Braga
- Centro de Biotecnologia, Universidade Federal da Paraíba João Pessoa, Brazil
| | - Eric Lazartigues
- Department of Pharmacology and Experimental Therapeutics and Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center New Orleans, LA, USA
| | | |
Collapse
|
32
|
Pereira TMC, Pimenta FS, Porto ML, Baldo MP, Campagnaro BP, Gava AL, Meyrelles SS, Vasquez EC. Coadjuvants in the Diabetic Complications: Nutraceuticals and Drugs with Pleiotropic Effects. Int J Mol Sci 2016; 17:ijms17081273. [PMID: 27527163 PMCID: PMC5000671 DOI: 10.3390/ijms17081273] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 07/27/2016] [Accepted: 07/29/2016] [Indexed: 12/19/2022] Open
Abstract
Because diabetes mellitus (DM) is a multifactorial metabolic disease, its prevention and treatment has been a constant challenge for basic and clinical investigators focused on translating their discoveries into clinical treatment of this complex disorder. In this review, we highlight recent experimental and clinical evidences of potential coadjuvants in the management of DM, such as polyphenols (quercetin, resveratrol and silymarin), cultured probiotic microorganisms and drugs acting through direct/indirect or pleiotropic effects on glycemic control in DM. Among several options, we highlight new promising therapeutic coadjuvants, including chemical scavengers, the probiotic kefir and the phosphodiesterase 5 inhibitors, which besides the reduction of hyperglycemia and ameliorate insulin resistance, they reduce oxidative stress and improve endothelial dysfunction in the systemic vascular circulation. In the near future, experimental studies are expected to clear the intracellular pathways involving coadjuvants. The design of clinical trials may also contribute to new strategies with coadjuvants against the harmful effects of diabetic complications.
Collapse
Affiliation(s)
- Thiago Melo Costa Pereira
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Av. Comissario Jose Dantas Melo 21, Boa Vista, 29102-920 Vila Velha, Brazil.
- Federal Institute of Education, Science and Technology (IFES), 29106-010 Vila Velha, Brazil.
| | - Fabio Silva Pimenta
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Av. Comissario Jose Dantas Melo 21, Boa Vista, 29102-920 Vila Velha, Brazil.
- Burn Treatment Center, Children State Hospital, 29056-030 Vitoria, Brazil.
| | - Marcella Lima Porto
- Federal Institute of Education, Science and Technology (IFES), 29106-010 Vila Velha, Brazil.
| | - Marcelo Perim Baldo
- Department of Pathophysiology, Montes Claros State University, 39401-089, Montes Claros, Brazil.
| | - Bianca Prandi Campagnaro
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Av. Comissario Jose Dantas Melo 21, Boa Vista, 29102-920 Vila Velha, Brazil.
| | - Agata Lages Gava
- Laboratory of Translational Physiology, Federal University of Espirito Santo (Ufes), 29047-100 Vitoria, Brazil.
- Division of Nephrology, McMaster University, Hamilton, ON L8N 4A6, Canada.
| | - Silvana Santos Meyrelles
- Laboratory of Translational Physiology, Federal University of Espirito Santo (Ufes), 29047-100 Vitoria, Brazil.
| | - Elisardo Corral Vasquez
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Av. Comissario Jose Dantas Melo 21, Boa Vista, 29102-920 Vila Velha, Brazil.
- Laboratory of Translational Physiology, Federal University of Espirito Santo (Ufes), 29047-100 Vitoria, Brazil.
| |
Collapse
|
33
|
Shen K, Johnson DW, Gobe GC. The role of cGMP and its signaling pathways in kidney disease. Am J Physiol Renal Physiol 2016; 311:F671-F681. [PMID: 27413196 DOI: 10.1152/ajprenal.00042.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 07/10/2016] [Indexed: 01/20/2023] Open
Abstract
Cyclic nucleotide signal transduction pathways are an emerging research field in kidney disease. Activated cell surface receptors transduce their signals via intracellular second messengers such as cAMP and cGMP. There is increasing evidence that regulation of the cGMP-cGMP-dependent protein kinase 1-phosphodiesterase (cGMP-cGK1-PDE) signaling pathway may be renoprotective. Selective PDE5 inhibitors have shown potential in treating kidney fibrosis in patients with chronic kidney disease (CKD), via their downstream signaling, and these inhibitors also have known activity as antithrombotic and anticancer agents. This review gives an outline of the cGMP-cGK1-PDE signaling pathways and details the downstream signaling and regulatory functions that are modulated by cGK1 and PDE inhibitors with regard to antifibrotic, antithrombotic, and antitumor activity. Current evidence that supports the renoprotective effects of regulating cGMP-cGK1-PDE signaling is also summarized. Finally, the effects of icariin, a natural plant extract with PDE5 inhibitory function, are discussed. We conclude that regulation of cGMP-cGK1-PDE signaling might provide novel, therapeutic strategies for the worsening global public health problem of CKD.
Collapse
Affiliation(s)
- Kunyu Shen
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Second School of Clinical Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China; and
| | - David W Johnson
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia; Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Glenda C Gobe
- Centre for Kidney Disease Research, School of Medicine, Translational Research Institute, The University of Queensland, Brisbane, Australia;
| |
Collapse
|
34
|
Bae HK, Lee H, Kim KC, Hong YM. The effect of sildenafil on right ventricular remodeling in a rat model of monocrotaline-induced right ventricular failure. KOREAN JOURNAL OF PEDIATRICS 2016; 59:262-70. [PMID: 27462355 PMCID: PMC4958704 DOI: 10.3345/kjp.2016.59.6.262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 04/25/2016] [Accepted: 05/10/2016] [Indexed: 11/29/2022]
Abstract
Purpose Pulmonary arterial hypertension (PAH) leads to right ventricular failure (RVF) as well as an increase in pulmonary vascular resistance. Our purpose was to study the effect of sildenafil on right ventricular remodeling in a rat model of monocrotaline (MCT)-induced RVF. Methods The rats were distributed randomly into 3 groups. The control (C) group, the monocrotaline (M) group (MCT 60 mg/kg) and the sildenafil (S) group (MCT 60 mg/kg+ sildenafil 30 mg/kg/day for 28 days). Masson Trichrome staining was used for heart tissues. Western blot analysis and immunohistochemical staining were performed. Results The mean right ventricular pressure (RVP) was significantly lower in the S group at weeks 1, 2, and 4. The number of intra-acinar arteries and the medial wall thickness of the pulmonary arterioles significantly lessened in the S group at week 4. The collagen content also decreased in heart tissues in the S group at week 4. Protein expression levels of B-cell lymphoma-2 (Bcl-2)-associated X, caspase-3, Bcl-2, interleukin (IL)-6, matrix metalloproteinase (MMP)-2, endothelial nitric oxide synthase (eNOS), endothelin (ET)-1 and ET receptor A (ERA) in lung tissues greatly decreased in the S group at week 4 according to immunohistochemical staining. According to Western blotting, protein expression levels of troponin I, brain natriuretic peptide, caspase-3, Bcl-2, tumor necrosis factor-α, IL-6, MMP-2, eNOS, ET-1, and ERA in heart tissues greatly diminished in the S group at week 4. Conclusion Sildenafil alleviated right ventricular hypertrophy and mean RVP. These data suggest that sildenafil improves right ventricular function.
Collapse
Affiliation(s)
- Hyun Kyung Bae
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| | - Hyeryon Lee
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| | - Kwan Chang Kim
- Department of Thoracic and Cardiovascular Surgery, Ewha Womans University School of Medicine, Seoul, Korea
| | - Young Mi Hong
- Department of Pediatrics, Ewha Womans University School of Medicine, Seoul, Korea
| |
Collapse
|
35
|
Bernardes FP, Batista AT, Porto ML, Vasquez EC, Campagnaro BP, Meyrelles SS. Protective effect of sildenafil on the genotoxicity and cytotoxicity in apolipoprotein E-deficient mice bone marrow cells. Lipids Health Dis 2016; 15:100. [PMID: 27229150 PMCID: PMC4882816 DOI: 10.1186/s12944-016-0268-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Accepted: 05/20/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The pharmacological inhibitor of phosphodiesterase 5 (PDE5), sildenafil, is a promising candidate for antioxidant therapy that can result in cardiovascular protection. In addition to its known effects on the cardiovascular system, hypercholesterolemia leads to increased oxidative stress and DNA damage in the bone marrow, which is a non-classical target organ of atherosclerosis. In the present study, we evaluate oxidative stress and assess the effect of genomic instability on cell cycle kinetics in atherosclerotic animals and determine if sildenafil reverses these detrimental effects in bone marrow cells. METHODS Experiments were performed in male wild-type (WT) and apolipoprotein E knockout mice (apoE(-/-)) (9 weeks of age). apoE(-/-) mice were randomly distributed into the following 2 groups: sildenafil-treated (40 mg/kg/day for 3 weeks, n = 8) and vehicle-treated (n = 8), by oral gavage. After treatment, bone marrow cells were isolated to assess the production of superoxide anions and hydrogen peroxide, determine cell cycle kinetics and evaluate the presence of micronucleated cells. RESULTS Sildenafil treatment reduced the cytoplasmic levels of superoxide anion (~95% decrease, p < 0.05) and decreased hydrogen peroxide (~30% decrease, p < 0.05). Moreover, we observed protective effects on the DNA of bone marrow cells, including normal cell cycling, decreased DNA fragmentation and a diminished frequency of micronucleated cells. CONCLUSION Our data reveal that the excessive production of ROS in atherosclerotic mice overcome the DNA repair pathways in bone marrow cells. The novelty of the present study is that the administration of sildenafil reduced ROS to baseline levels and, consequently, reverted the DNA damage and its outcomes in bone marrow cells.
Collapse
Affiliation(s)
- Franciane P Bernardes
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Alan T Batista
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
| | - Marcella L Porto
- Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil
| | - Elisardo C Vasquez
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil
| | - Bianca P Campagnaro
- Pharmaceutical Sciences Graduate Program, Vila Velha University (UVV), Vila Velha, ES, Brazil
| | - Silvana S Meyrelles
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, Brazil.
| |
Collapse
|
36
|
Cavalcanti CDO, Alves RR, de Oliveira AL, Cruz JDC, de França-Silva MDS, Braga VDA, Balarini CDM. Inhibition of PDE5 Restores Depressed Baroreflex Sensitivity in Renovascular Hypertensive Rats. Front Physiol 2016; 7:15. [PMID: 26858657 PMCID: PMC4729906 DOI: 10.3389/fphys.2016.00015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2015] [Accepted: 01/11/2016] [Indexed: 01/05/2023] Open
Abstract
Renal artery stenosis is frequently associated with resistant hypertension, which is defined as failure to normalize blood pressure (BP) even when combined drugs are used. Inhibition of PDE5 by sildenafil has been shown to increase endothelial function and decrease blood pressure in experimental models. However, no available study evaluated the baroreflex sensitivity nor autonomic balance in renovascular hypertensive rats treated with sildenafil. In a translational medicine perspective, our hypothesis is that sildenafil could improve autonomic imbalance and baroreflex sensitivity, contributing to lower blood pressure. Renovascular hypertensive 2-kidney-1-clip (2K1C) and sham rats were treated with sildenafil (45 mg/Kg/day) during 7 days. At the end of treatment, BP and heart rate (HR) were recorded in conscious rats after a 24-h-recovery period. Spontaneous and drug-induced baroreflex sensitivity and autonomic tone were evaluated; in addition, lipid peroxidation was measured in plasma samples. Treatment was efficient in increasing both spontaneous and induced baroreflex sensitivity in treated hypertensive animals. Inhibition of PDE5 was also capable of ameliorating autonomic imbalance in 2K1C rats and decreasing systemic oxidative stress. Taken together, these beneficial effects resulted in significant reductions in BP without affecting HR. We suggest that sildenafil could be considered as a promising alternative to treat resistant hypertension.
Collapse
Affiliation(s)
| | - Rafael R Alves
- Centro de Ciências Médicas, Universidade Federal da ParaíbaJoao Pessoa, Brazil; Centro de Ciências da Saúde, Universidade Federal da ParaíbaJoao Pessoa, Brazil
| | - Alessandro L de Oliveira
- Centro de Ciências Médicas, Universidade Federal da ParaíbaJoao Pessoa, Brazil; Centro de Ciências da Saúde, Universidade Federal da ParaíbaJoao Pessoa, Brazil
| | | | | | | | - Camille de Moura Balarini
- Centro de Biotecnologia, Universidade Federal da ParaíbaJoao Pessoa, Brazil; Centro de Ciências da Saúde, Universidade Federal da ParaíbaJoao Pessoa, Brazil
| |
Collapse
|
37
|
Friques AGF, Arpini CM, Kalil IC, Gava AL, Leal MA, Porto ML, Nogueira BV, Dias AT, Andrade TU, Pereira TMC, Meyrelles SS, Campagnaro BP, Vasquez EC. Chronic administration of the probiotic kefir improves the endothelial function in spontaneously hypertensive rats. J Transl Med 2015; 13:390. [PMID: 26715471 PMCID: PMC4696190 DOI: 10.1186/s12967-015-0759-7] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 12/22/2015] [Indexed: 12/13/2022] Open
Abstract
Background The beverage obtained by fermentation of milk with kefir grains, a complex matrix containing acid bacteria and yeasts, has been shown to have beneficial effects in various diseases. However, its effects on hypertension and endothelial dysfunction are not yet clear. In this study, we evaluated the effects of kefir on endothelial cells and vascular responsiveness in spontaneously hypertensive rats (SHR). Methods SHR were treated with kefir (0.3 mL/100 g body weight) for 7, 15, 30 and 60 days and compared with non-treated SHR and with normotensive Wistar-Kyoto rats. Vascular endothelial function was evaluated in aortic rings through the relaxation response to acetylcholine (ACh). The balance between reactive oxygen species (ROS) and nitric oxide (NO) synthase was evaluated through specific blockers in the ACh-induced responses and through flow cytometry in vascular tissue. Results Significant effects of kefir were observed only after treatment for 60 days. The high blood pressure and tachycardia exhibited by the SHR were attenuated by approximately 15 % in the SHR-kefir group. The impaired ACh-induced relaxation of the aortic rings observed in the SHR (37 ± 4 %, compared to the Wistar rats: 74 ± 5 %), was significantly attenuated in the SHR group chronically treated with kefir (52 ± 4 %). The difference in the area under the curve between before and after the NADPH oxidase blockade or NO synthase blockade of aortic rings from SHR were of approximately +90 and −60 %, respectively, when compared with Wistar rats. In the aortic rings from the SHR-kefir group, these values were reduced to +50 and −40 %, respectively. Flow cytometric analysis of aortic endothelial cells revealed increased ROS production and decreased NO bioavailability in the SHR, which were significantly attenuated by the treatment with kefir. Scanning electronic microscopy showed vascular endothelial surface injury in SHR, which was partially protected following administration of kefir for 60 days. In addition, the recruitment of endothelial progenitor cells was decreased in the non-treated SHR and partially restored by kefir treatment. Conclusions Kefir treatment for 60 days was able to improve the endothelial function in SHR by partially restoring the ROS/NO imbalance and the endothelial architecture due to endothelial progenitor cells recruitment.
Collapse
Affiliation(s)
- Andreia G F Friques
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil.
| | - Clarisse M Arpini
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil.
| | - Ieda C Kalil
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil.
| | - Agata L Gava
- Division of Nephrology, McMaster University, Hamilton, ON, Canada.
| | - Marcos A Leal
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Marcella L Porto
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil. .,Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil.
| | - Breno V Nogueira
- Department of Morphology, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Ananda T Dias
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Tadeu U Andrade
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil.
| | - Thiago Melo C Pereira
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil. .,Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil.
| | - Silvana S Meyrelles
- Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| | - Bianca P Campagnaro
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil.
| | - Elisardo C Vasquez
- Pharmaceutical Sciences Graduate Program, Vila Velha University, Vila Velha, ES, Brazil. .,Laboratory of Translational Physiology, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| |
Collapse
|
38
|
Liu KL, Canaple L, Del Carmine P, Gauthier K, Beylot M, Lo M. Thyroid hormone receptor-α deletion decreases heart function and exercise performance in apolipoprotein E-deficient mice. Physiol Genomics 2015; 48:73-81. [PMID: 26672044 DOI: 10.1152/physiolgenomics.00115.2015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/14/2015] [Indexed: 11/22/2022] Open
Abstract
The deletion of thyroid hormone receptor-α (TRα) in atherosclerosis-prone apolipoprotein E-deficient (ApoE(-/-)) mice (ApoE(-/-)TRα(0/0)) accelerates the formation of atherosclerotic plaques without aggravation of hypercholesterolemia. To evaluate other predisposition risk factors to atherosclerosis in this model, we studied blood pressure (BP) and cardiac and vascular functions, as well as exercise tolerance in young adult ApoE(-/-)TRα(0/0) mice before the development of atherosclerotic plaques. Telemetric BP recorded for 4 consecutive days showed that the spontaneous systolic BP was slightly decreased in ApoE(-/-)TRα(0/0) compared with ApoE(-/-) mice associated with a reduced locomotor activity. The percentage of animals that completed endurance (57% vs. 89%) and maximal running (0% vs. 89% at 46 cm/s speed in ApoE(-/-)TRα(0/0) and ApoE(-/-) mice, respectively) tests was lower in ApoE(-/-)TRα(0/0) mice. Moreover, during the maximal running test, both maximal running speed and running distance were significantly reduced in ApoE(-/-)TRα(0/0) mice, associated with a blunted BP response to exercise. Transthoracic echocardiography revealed a decreased interventricular septum thickness and an increased end-systolic left ventricular volume in ApoE(-/-)TRα(0/0) mice. Accordingly, left ventricular fractional shortening, ejection fraction, and stroke volume were all significantly decreased in ApoE(-/-)TRα(0/0) mice with a concomitant blunted cardiac output. No interstrain difference was observed in vascular reactivity, except that ApoE(-/-)TRα(0/0) mice exhibited an enhanced acetylcholine-induced relaxation in mesenteric and distal femoral arteries. In conclusion, the deletion of TRα in ApoE(-/-) mice alters cardiac structure and contractility; both could contribute to blunted BP response to physical exercise and impaired exercise performance.
Collapse
Affiliation(s)
- Kiao Ling Liu
- Neurocardiology Unit - EA 4612, Institute of Pharmaceutical and Biological Sciences, University Claude Bernard Lyon 1, Lyon, France;
| | - Laurence Canaple
- Institute of Functional Genomics of Lyon, University Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; and
| | - Peggy Del Carmine
- Technical Platform ANIPHY, University Claude Bernard Lyon 1, Lyon, France
| | - Karine Gauthier
- Institute of Functional Genomics of Lyon, University Claude Bernard Lyon 1, Centre National de la Recherche Scientifique, Ecole Normale Supérieure de Lyon, Lyon, France; and
| | - Michel Beylot
- Neurocardiology Unit - EA 4612, Institute of Pharmaceutical and Biological Sciences, University Claude Bernard Lyon 1, Lyon, France; Technical Platform ANIPHY, University Claude Bernard Lyon 1, Lyon, France
| | - Ming Lo
- Neurocardiology Unit - EA 4612, Institute of Pharmaceutical and Biological Sciences, University Claude Bernard Lyon 1, Lyon, France
| |
Collapse
|
39
|
Freitas Lima LC, Braga VDA, do Socorro de França Silva M, Cruz JDC, Sousa Santos SH, de Oliveira Monteiro MM, Balarini CDM. Adipokines, diabetes and atherosclerosis: an inflammatory association. Front Physiol 2015; 6:304. [PMID: 26578976 PMCID: PMC4630286 DOI: 10.3389/fphys.2015.00304] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 10/12/2015] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases can be considered the most important cause of death in diabetic population and diabetes can in turn increase the risk of cardiovascular events. Inflammation process is currently recognized as responsible for the development and maintenance of diverse chronic diseases, including diabetes and atherosclerosis. Considering that adipose tissue is an important source of adipokines, which may present anti and proinflammatory effects, the aim of this review is to explore the role of the main adipokines in the pathophysiology of diabetes and atherosclerosis, highlighting the therapeutic options that could arise from the manipulation of these signaling pathways both in humans and in translational models.
Collapse
Affiliation(s)
| | - Valdir de Andrade Braga
- Biotechnology Center, Federal University of Paraiba (Universidade Federal da Paraíba)Joao Pessoa, Brazil
| | | | - Josiane de Campos Cruz
- Biotechnology Center, Federal University of Paraiba (Universidade Federal da Paraíba)Joao Pessoa, Brazil
| | - Sérgio H. Sousa Santos
- Biological Sciences Institute, Federal University of Minas GeraisBelo Horizonte, Brazil
- Health Science Post-Graduate Program, State University of Montes ClarosMontes Claros, Brazil
| | | | - Camille de Moura Balarini
- Biotechnology Center, Federal University of Paraiba (Universidade Federal da Paraíba)Joao Pessoa, Brazil
- Health Sciences Center, Federal University of Paraiba (Universidade Federal da Paraíba)Joao Pessoa, Brazil
| |
Collapse
|
40
|
Peixoto CA, Gomes FODS. The role of phosphodiesterase-5 inhibitors in prostatic inflammation: a review. JOURNAL OF INFLAMMATION-LONDON 2015; 12:54. [PMID: 26379476 PMCID: PMC4570643 DOI: 10.1186/s12950-015-0099-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 08/31/2015] [Indexed: 12/18/2022]
Abstract
Clinical and basic experimental evidence indicates that chronic inflammation is the greatest factor in benign prostatic hyperplasia (BPH) progression, which is the most common cause of Lower Urinary Tract Symptoms (LUTS). The use of anti-inflammatory agents such as steroids, cyclooxygenase-2 (COX-2) and phytotherapics have been investigated as forms of treatment for various prostate diseases. Recent evidence has demonstrated that PDE5 inhibitors (PDE5Is) improve symptoms of BPH/LUTS, possibly as a result of the relaxing of the smooth muscle fibers of the bladder and prostate by NO/cGMPc signaling, or by improving RhoA/Rho-kinase (ROCK), and reduction of the hyperactivity of the autonomic nervous system. However, some results have suggested that besides vasodilatation and their anti-proliferative effect, PDE5Is exert a direct anti-inflammatory effect, by raising cGMP. Given that inflammation is major factor in benign prostatic hyperplasia (BPH) progression, PDE5Is could act also restore prostatic function as they act as potent anti-inflammatory drugs. This review aims to provide a comprehensive summary of the use of phosphodiesterase-5 inhibitors to treat prostatic inflammation.
Collapse
Affiliation(s)
- Christina Alves Peixoto
- Laboratório de Ultraestrutura, Centro de Pesquisas Aggeu Magalhães (CPqAM-FIOCRUZ), Fundação Oswaldo Cruz, Av. Moraes Rego s/n, CEP: 50670-420, Cidade Universitária, Recife, PE Brazil
| | - Fabiana Oliveira Dos Santos Gomes
- Laboratório de Ultraestrutura, Centro de Pesquisas Aggeu Magalhães (CPqAM-FIOCRUZ), Fundação Oswaldo Cruz, Av. Moraes Rego s/n, CEP: 50670-420, Cidade Universitária, Recife, PE Brazil
| |
Collapse
|
41
|
Ziqiang X, Jingjun W, Jianjian Z, Yong L, Peng X. Tadalafil attenuates graft arteriosclerosis of aortic transplant in a rat model. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2015; 18:927-31. [PMID: 26526520 PMCID: PMC4620194] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
OBJECTIVES Tadalafil can restore endothelial function and treat atherosclerosis. However, the effect of tadalafil on transplant arteriosclerosis remains unclear. In this study, we explore the effects of tadalafil on allograft vasculopathy. MATERIALS AND METHODS Male Brow-Norway rats supplied aorta grafts for Male Lewis rats. All recipients were divided into 3 groups: saline as placebo (control) treated group, low dose tadalafil (0.5 mg/kg/day) treated group, and high dose tadalafil (1.0 mg/kg/day) treated group. Eight weeks after transplantation, the grafts were harvested at and analyzed by histological and Western blot analysis. An enzyme-linked immunosorbent assay (ELISA) was used for measure of plasma cyclic guanylate monophosphate (cGMP). RESULTS the treatment with tadalafil significantly alleviated the neointimal thickness of aortas compared with the control group (P<0.05). Tadalafil also remarkably enhanced the production of cGMP in plasma and expression of cGMP-dependent kinase I (PKG-I) and RhoA compared with control group (P<0.05). CONCLUSION These results showed that tadalafil can attenuate graft arteriosclerosis by cGMP -PKG-I pathway.
Collapse
Affiliation(s)
- Xu Ziqiang
- Transplantation Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wang Jingjun
- Transplantation Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zheng Jianjian
- Transplantation Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liang Yong
- Transplantation Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xia Peng
- Transplantation Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China,Corresponding author: Xia Peng, Transplantation Center, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China. Tel: 86+0577-55579473;
| |
Collapse
|
42
|
Abstract
The view of atherosclerosis as an inflammatory disease has emerged from observations of immune activation and inflammatory signalling in human atherosclerotic lesions, from the definition of inflammatory biomarkers as independent risk factors for cardiovascular events, and from evidence of low-density lipoprotein-induced immune activation. Studies in animal models of hyperlipidaemia have also supported the beneficial effects of countering inflammation to delay atherosclerosis progression. Specific inflammatory pathways with relevance to human diseases have been identified, and inhibitors of these pathways are either already in use for the treatment of other diseases, or are under development and evaluation. These include 'classic' drugs (such as allopurinol, colchicine, and methotrexate), biologic therapies (for example tumour necrosis factor inhibitors and IL-1 neutralization), as well as targeting of lipid mediators (such as phospholipase inhibitors and antileukotrienes) or intracellular pathways (inhibition of NADPH oxidase, p38 mitogen-activated protein kinase, or phosphodiesterase). The evidence supporting the use of anti-inflammatory therapies for atherosclerosis is mainly based on either observational or small interventional studies evaluating surrogate markers of disease activity. Nevertheless, these data are crucial to understand the role of inflammation in atherosclerosis, and to design randomized controlled studies to evaluate the effect of specific anti-inflammatory strategies on cardiovascular outcomes.
Collapse
Affiliation(s)
- Magnus Bäck
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, L8:03, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| | - Göran K Hansson
- Experimental Cardiovascular Research Unit, Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, L8:03, Karolinska University Hospital, SE-171 76 Stockholm, Sweden
| |
Collapse
|
43
|
High fat diet exacerbates vascular endothelial dysfunction in rats exposed to continuous hypobaric hypoxia. Biochem Biophys Res Commun 2015; 457:485-91. [PMID: 25603049 DOI: 10.1016/j.bbrc.2015.01.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 01/10/2015] [Indexed: 01/28/2023]
Abstract
Independently, a high fat diet and hypoxia are associated with vascular endothelial dysfunction (VED) and often occur concurrently in patients. Nevertheless, the effects of a high fat diet on vascular endothelial function combined with hypoxia, a situation occurring with increasing frequency in many parts of the world, remain largely unknown. We investigated the effects of a high fat diet on vascular endothelial function in rats exposed to continuous hypoxia for 4 weeks. Seventy two male Sprague-Dawley rats were randomly divided into 3 groups: a hypoxia group fed regular chow, a combined hypoxia and high fat diet (HFD) group, and for comparison, rats maintained in normoxia, regular chow conditions were set as baseline (BL) group. The experimental data of BL group were obtained at beginning of hypoxia given in the other groups. Continuous hypoxia was induced in a hypobaric chamber maintained at an altitude of 5000 m. Compared to hypoxic conditions alone, hypoxia plus a HFD prevented adaptive changes in plasma nitric oxide (NOx) levels and caused earlier and more severe changes in aortic endothelial structures. Functionally, hypoxia plus a HFD resulted in impaired endothelium-dependent vasorelaxation responses to acetylcholine and altered the bioavailability of the nitric oxide synthase (NOS) substrate L-Arginine. At the molecular level, hypoxia plus a HFD blunted increases in endothelial NOS (eNOS) mRNA and protein in aortic endothelial tissue. Taken together, our findings demonstrate that in the setting of hypoxia, a high fat diet leads to earlier and more severe VED than hypoxia alone. These data have important implications for populations residing at high-altitude, as dietary patterns shift towards increased fat intake.
Collapse
|
44
|
Rua EDAO, Porto ML, Ramos JPL, Nogueira BV, Meyrelles SS, Vasquez EC, Pereira TC. Effects of tobacco smoking during pregnancy on oxidative stress in the umbilical cord and mononuclear blood cells of neonates. J Biomed Sci 2014; 21:105. [PMID: 25547987 PMCID: PMC4302517 DOI: 10.1186/s12929-014-0105-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 12/05/2014] [Indexed: 12/21/2022] Open
Abstract
Background Although cigarette smoke is known to be a complex mixture of over 4000 substances that can lead to damage through active or passive smoking, its mechanisms and biochemical consequences in pregnancy and neonates are not yet fully understood. Therefore, in the present study, we propose to study the impact of smoking during gestation on the viability of blood mononuclear cells (MNC) from umbilical cords of newborns to assess the degree of oxidative stress and cell viability. After childbirth, the cord blood and the umbilical cord were immediately collected in public hospitals in Greater Vitoria, ES, Brazil. Flow cytometry was used to analyze the cord blood followed by biochemical and histological tests to analyze possible changes in the umbilical cord. Results Pregnant smokers had a reduction of MNC viability from the umbilical cord (10%), an increase in the production of reactive oxygen species (ROS) and an increase in cell apoptosis (~2-fold) compared to pregnant non-smokers. In the umbilical cord, it was observed an increase of advanced oxidation protein products - AOPP (~2.5-fold) and a loss of the typical architecture and disposition of endothelial cells from the umbilical artery. Conclusions These data suggest that maternal cigarette smoking during pregnancy (even in small amounts) may compromise the viability of MNC cells and damage the umbilical cord structure, possibly by excessive ROS bioavailability.
Collapse
|
45
|
Carneiro SS, Carminati RZ, Freitas FPS, Podratz PL, Balarini CM, Graceli JB, Meyrelles SS, Vasquez EC, Gava AL. Endogenous female sex hormones delay the development of renal dysfunction in apolipoprotein E-deficient mice. Lipids Health Dis 2014; 13:176. [PMID: 25422135 PMCID: PMC4280709 DOI: 10.1186/1476-511x-13-176] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Accepted: 11/15/2014] [Indexed: 12/16/2022] Open
Abstract
Background Hypercholesterolemia is a well-established risk factor for the development of kidney injury. Considering that female sex hormones may play a preventative role in both cardiovascular and renal diseases, the aim of the present study was to evaluate the effects of female sex hormones on hypercholesterolemia-induced renal dysfunction. Methods Apolipoprotein E-deficient (ApoE) and C57 control female mice underwent an ovariectomy (OVX) or sham surgery and after 2 months, creatinine clearance, uremia and proteinuria were determined. Renal oxidative stress and lipid deposition were also quantified. Values are presented as mean ± SEM. Statistical analyses were performed using Two-way ANOVA followed by Tukey’s post hoc test. Results Creatinine clearance (μL/min) was similar between C57 (171 ± 17) and ApoE (140 ± 26) mice underwent sham surgery. OVX resulted in a reduced glomerular filtration rate in both C57 (112 ± 8, ~ − 35%, p < 0.05) and ApoE (61 ± 10, ~ − 56%, p < 0.05) animals. Plasma levels of urea (mg/dL) were higher in both ApoE groups (Sham: 73 ± 7; OVX: 73 ± 8, p < 0.05) when compared to C57 animals (Sham: 49 ± 3; OVX: 60 ± 4), with no changes among ovariectomized groups. Proteinuria levels (mg/24 h) were similar between C57 (Sham: 25.1 ± 5.7; OVX: 33.7 ± 4.7) and ApoE sham animals (26.4 ± 3.5), however, 24-h urine protein excretion was augmented in ApoE OVX animals (49.6 ± 5.8, p < 0.05). Histological kidney analysis demonstrated that the absence of female sex hormones resulted in increased oxidative stress, which was more severe in ApoE mice (C57 Sham: 9.2 ± 0.4; C57 OVX: 22.9 ± 1.0; ApoE Sham: 13.9 ± 0.7; ApoE OVX: 34.0 ± 1.4 au x 103, p < 0.05). As expected, ApoE mice presented higher lipid deposition, which was not affected by OVX (C57 Sham: 0 ± 0; C57 OVX: 0 ± 0; ApoE Sham: 6.8 ± 1.6; ApoE OVX: 5.2 ± 0.8% x 10−2, p < 0.05). Ovariectomy resulted in a similar reduction in ER-α protein expression in the renal cortex (C57: 0.78 ± 0.04; ApoE: 0.81 ± 0.04 au, p < 0.05) when compared to sham animals (C57:1.00 ± 0.04; ApoE: 1.03 ± 0.03 au). Conclusion Taken together these data indicate that female sex hormones may delay hypercholesterolemia-induced renal dysfunction and emphasizes the importance of plasma cholesterol control in post-menopausal women.
Collapse
Affiliation(s)
| | | | | | | | | | - Jones B Graceli
- Laboratory of Translational Physiology, Physiological Sciences Graduate Program, Health Sciences Center, Federal University of Espirito Santo, Av Marechal Campos 1468, 29042-755 Vitoria, ES, Brazil.
| | | | | | | |
Collapse
|
46
|
Dias AT, Cintra AS, Frossard JC, Palomino Z, Casarini DE, Gomes IBS, Balarini CM, Gava AL, Campagnaro BP, Pereira TMC, Meyrelles SS, Vasquez EC. Inhibition of phosphodiesterase 5 restores endothelial function in renovascular hypertension. J Transl Med 2014; 12:250. [PMID: 25223948 PMCID: PMC4172908 DOI: 10.1186/s12967-014-0250-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 08/30/2014] [Indexed: 01/25/2023] Open
Abstract
Background The clipping of an artery supplying one of the two kidneys (2K1C) activates the renin-angiotensin (Ang) system (RAS), resulting in hypertension and endothelial dysfunction. Recently, we demonstrated the intrarenal beneficial effects of sildenafil on the high levels of Ang II and reactive oxygen species (ROS) and on high blood pressure (BP) in 2K1C mice. Thus, in the present study, we tested the hypothesis that sildenafil improves endothelial function in hypertensive 2K1C mice by improving the NO/ROS balance. Methods 2K1C hypertension was induced in C57BL/6 mice. Two weeks later, they were treated with sildenafil (40 mg/kg/day, via oral) or vehicle for 2 weeks and compared with sham mice. At the end of the treatment, the levels of plasma and intrarenal Ang peptides were measured. Endothelial function and ROS production were assessed in mesenteric arterial bed (MAB). Results The 2K1C mice exhibited normal plasma levels of Ang I, II and 1–7, whereas the intrarenal Ang I and II were increased (~35% and ~140%) compared with the Sham mice. Sildenafil normalized the intrarenal Ang I and II and increased the plasma (~45%) and intrarenal (+15%) Ang 1–7. The 2K1C mice exhibited endothelial dysfunction, primarily due to increased ROS and decreased NO productions by endothelial cells, which were ameliorated by treatment with sildenafil. Conclusion These data suggest that the effects of sildenafil on endothelial dysfunction in 2K1C mice may be due to interaction with RAS and restoring NO/ROS balance in the endothelial cells from MAB. Thus, sildenafil is a promising candidate drug for the treatment of hypertension accompanied by endothelial dysfunction and kidney disease.
Collapse
|
47
|
Dias AT, Rodrigues BP, Porto ML, Gava AL, Balarini CM, Freitas FPS, Palomino Z, Casarini DE, Campagnaro BP, Pereira TMC, Meyrelles SS, Vasquez EC. Sildenafil ameliorates oxidative stress and DNA damage in the stenotic kidneys in mice with renovascular hypertension. J Transl Med 2014; 12:35. [PMID: 24502628 PMCID: PMC3922021 DOI: 10.1186/1479-5876-12-35] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Accepted: 02/04/2014] [Indexed: 12/01/2022] Open
Abstract
Background Oxidative stress and DNA damage have been implicated in the pathogenesis of renovascular hypertension induced by renal artery stenosis in the two-kidney, one-clip (2K1C) Goldblatt model. Considering our previous report indicating that the chronic blockade of phosphodiesterase 5 with sildenafil (Viagra®) has marked beneficial effects on oxidative stress and DNA damage, we tested the hypothesis that sildenafil could also protect the stenotic kidneys of 2K1C hypertensive mice against oxidative stress and genotoxicity. Methods The experiments were performed with C57BL6 mice subjected to renovascular hypertension by left renal artery clipping. Two weeks after clipping, the mice were treated with sildenafil (40 mg/kg/day for 2 weeks, 2K1C-sildenafil group) or the vehicle (2K1C). These mice were compared with control mice not subjected to renal artery clipping (Sham). After hemodynamic measurements, the stenotic kidneys were assessed using flow cytometry to evaluate cell viability and the comet assay to evaluate DNA damage. Measurements of intracellular superoxide anions and hydrogen peroxide levels as well as nitric oxide bioavailability were also obtained. Results Sildenafil treatment significantly reduced mean arterial pressure (15%), heart rate (8%), intrarenal angiotensin II (50%) and renal atrophy (36%). In addition, it caused a remarkable decrease of reactive oxygen species production. On the other hand, sildenafil increased nitric oxide levels relative to those in the nontreated 2K1C mice. Sildenafil treatment also significantly reduced the high level of kidney DNA damage that is a characteristic of renovascular hypertensive mice. Conclusions Our data reveal that sildenafil has a protective effect on the stenotic kidneys of 2K1C mice, suggesting a new use of phosphodiesterase 5 inhibitors for protection against the DNA damage observed in the hypoperfused kidneys of individuals with renovascular hypertension. Further translational research is necessary to delineate the mechanisms involved in the prevention of renal stenosis in the clinical setting.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Elisardo C Vasquez
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo, Vitoria, ES, Brazil.
| |
Collapse
|
48
|
Chen W, Oberwinkler H, Werner F, Gaßner B, Nakagawa H, Feil R, Hofmann F, Schlossmann J, Dietrich A, Gudermann T, Nishida M, Del Galdo S, Wieland T, Kuhn M. Atrial Natriuretic Peptide–Mediated Inhibition of Microcirculatory Endothelial Ca
2+
and Permeability Response to Histamine Involves cGMP-Dependent Protein Kinase I and TRPC6 Channels. Arterioscler Thromb Vasc Biol 2013; 33:2121-9. [DOI: 10.1161/atvbaha.113.001974] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Wen Chen
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Heike Oberwinkler
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Franziska Werner
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Birgit Gaßner
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Hitoshi Nakagawa
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Robert Feil
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Franz Hofmann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Jens Schlossmann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Alexander Dietrich
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Thomas Gudermann
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Motohiro Nishida
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Sabrina Del Galdo
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Thomas Wieland
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| | - Michaela Kuhn
- From the Institute of Physiology, University of Würzburg, Würzburg, Germany (W.C., H.O., F.W., B.G., H.N., M.K.); Interfakultäres Institut für Biochemie, University of Tübingen, Tübingen, Germany (R.F.); FOR 923, Technical University München, Garching, Germany (F.H.); Institute of Pharmacology and Toxicology, University of Regensburg, Regensburg, Germany (J.S.); Walther-Straub-Institute for Pharmacology and Toxicology, Ludwig-Maximilians-University München, Munich, Germany (A.D., T.G.); Department
| |
Collapse
|
49
|
Rodrigues BP, Campagnaro BP, Balarini CM, Pereira TMC, Meyrelles SS, Vasquez EC. Sildenafil ameliorates biomarkers of genotoxicity in an experimental model of spontaneous atherosclerosis. Lipids Health Dis 2013; 12:128. [PMID: 23981672 PMCID: PMC3766097 DOI: 10.1186/1476-511x-12-128] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2013] [Accepted: 08/23/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND It is well known that enhanced production of reactive oxygen species (ROS) leads to oxidative stress observed in atherosclerosis and that ROS can also cause damage in cellular macromolecules, including DNA. Considering previous report that sildenafil, an inhibitor of phosphodiesterase 5 (PDE5), has antioxidant effects, in the present study we evaluated the effect of this drug on genotoxicity of blood mononuclear cells (MNC) and liver cells from atherosclerotic apolipoprotein E knockout mice (apoE(-/-)). METHODS ROS production in MNC was evaluated by flow cytometry with the fluorescent dye dihydroethidium (DHE), a method that has been used to quantify the production of superoxide anion, and DNA damage was evaluated in both MNC and liver cells using the alkaline comet assay. Sildenafil-administered apoE(-/-) mice were compared with strain-matched mice administered with vehicle and with C57BL/6 wild-type (WT) mice. RESULTS MNC from apoE(-/-) vehicle exhibited a 2-fold increase in production of superoxide anion in comparison with WT. In contrast, sildenafil-administered apoE(-/-) mice showed superoxide anion levels similar to those observed in WT mice. Similarly, MNC and liver cells from apoE(-/-) vehicle mice showed a 4-fold and 2-fold augmented DNA fragmentation compared with WT, respectively, and sildenafil-administered apoE(-/-) mice exhibited minimal DNA damage in those cells similar to WT mice. CONCLUSIONS ApoE(-/-) mice chronically administered with sildenafil exhibited reduced levels of superoxide anion in MNC and less DNA fragmentation in MNC and liver cells, which are biomarkers of genotoxicity. Therefore, sildenafil may offer a new perspective to the use of PDE5 inhibitors to protect against DNA damage, in cells involved in the inflammatory and dyslipidemic processes that accompany atherosclerosis.
Collapse
Affiliation(s)
- Bianca P Rodrigues
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
| | - Bianca P Campagnaro
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
- Pharmaceutical Sciences Graduate Program, University of Vila Velha (UVV), Vila Velha, ES, Brazil
| | - Camille M Balarini
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
| | - Thiago M C Pereira
- Pharmaceutical Sciences Graduate Program, University of Vila Velha (UVV), Vila Velha, ES, Brazil
- Federal Institute of Education, Science and Technology (IFES), Vila Velha, ES, Brazil
| | - Silvana S Meyrelles
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
| | - Elisardo C Vasquez
- Laboratory of Translational Physiology, Health Sciences Center, Federal University of Espirito Santo (UFES), Vitoria, Brazil
- Pharmaceutical Sciences Graduate Program, University of Vila Velha (UVV), Vila Velha, ES, Brazil
- Emescam School of Health Sciences, Vitoria, Brazil
| |
Collapse
|
50
|
Chaumais MC, Perrin S, Sitbon O, Simonneau G, Humbert M, Montani D. Pharmacokinetic evaluation of sildenafil as a pulmonary hypertension treatment. Expert Opin Drug Metab Toxicol 2013; 9:1193-205. [DOI: 10.1517/17425255.2013.804063] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Marie-Camille Chaumais
- Université Paris-Sud, Faculté de Pharmacie,
Chatenay-Malabry, France
- AP-HP, Service de Pharmacie, DHU Thorax Innovation, Hôpital Antoine Béclère,
Clamart, France
- INSERM UMR 999, LabEx LERMIT, DHU Thorax Innovation, Centre Chirurgical Marie Lannelongue,
Le Plessis Robinson, France
| | - Swanny Perrin
- INSERM UMR 999, LabEx LERMIT, DHU Thorax Innovation, Centre Chirurgical Marie Lannelongue,
Le Plessis Robinson, France
- Université Paris-Sud, Faculté de Médecine,
Kremlin-Bicêtre, France
- AP-HP, Centre de Référence de l’Hypertension Pulmonaire Sévère, Service de Pneumologie et Réanimation Respiratoire, DHU Thorax Innovation, Hôpital de Bicêtre,
78, rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France .
| | - Olivier Sitbon
- INSERM UMR 999, LabEx LERMIT, DHU Thorax Innovation, Centre Chirurgical Marie Lannelongue,
Le Plessis Robinson, France
- Université Paris-Sud, Faculté de Médecine,
Kremlin-Bicêtre, France
- AP-HP, Centre de Référence de l’Hypertension Pulmonaire Sévère, Service de Pneumologie et Réanimation Respiratoire, DHU Thorax Innovation, Hôpital de Bicêtre,
78, rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France .
| | - Gérald Simonneau
- INSERM UMR 999, LabEx LERMIT, DHU Thorax Innovation, Centre Chirurgical Marie Lannelongue,
Le Plessis Robinson, France
- Université Paris-Sud, Faculté de Médecine,
Kremlin-Bicêtre, France
- AP-HP, Centre de Référence de l’Hypertension Pulmonaire Sévère, Service de Pneumologie et Réanimation Respiratoire, DHU Thorax Innovation, Hôpital de Bicêtre,
78, rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France .
| | - Marc Humbert
- INSERM UMR 999, LabEx LERMIT, DHU Thorax Innovation, Centre Chirurgical Marie Lannelongue,
Le Plessis Robinson, France
- Université Paris-Sud, Faculté de Médecine,
Kremlin-Bicêtre, France
- AP-HP, Centre de Référence de l’Hypertension Pulmonaire Sévère, Service de Pneumologie et Réanimation Respiratoire, DHU Thorax Innovation, Hôpital de Bicêtre,
78, rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France .
| | - David Montani
- INSERM UMR 999, LabEx LERMIT, DHU Thorax Innovation, Centre Chirurgical Marie Lannelongue,
Le Plessis Robinson, France
- Université Paris-Sud, Faculté de Médecine,
Kremlin-Bicêtre, France
- AP-HP, Centre de Référence de l’Hypertension Pulmonaire Sévère, Service de Pneumologie et Réanimation Respiratoire, DHU Thorax Innovation, Hôpital de Bicêtre,
78, rue du Général Leclerc, 94270 Le Kremlin-Bicêtre, France .
| |
Collapse
|