1
|
Chennupati R, Solga I, Wischmann P, Dahlmann P, Celik FG, Pacht D, Şahin A, Yogathasan V, Hosen MR, Gerdes N, Kelm M, Jung C. Chronic anemia is associated with systemic endothelial dysfunction. Front Cardiovasc Med 2023; 10:1099069. [PMID: 37234375 PMCID: PMC10205985 DOI: 10.3389/fcvm.2023.1099069] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/21/2023] [Indexed: 05/27/2023] Open
Abstract
Background In acute myocardial infarction and heart failure, anemia is associated with adverse clinical outcomes. Endothelial dysfunction (ED) is characterized by attenuated nitric oxide (NO)-mediated relaxation responses which is poorly studied in chronic anemia (CA). We hypothesized that CA is associated with ED due to increased oxidative stress in the endothelium. Methods CA was induced by repeated blood withdrawal in male C57BL/6J mice. Flow-Mediated Dilation (FMD) responses were assessed in CA mice using ultrasound-guided femoral transient ischemia model. Tissue organ bath was used to assess vascular responsiveness of aortic rings from CA mice, and in aortic rings incubated with red blood cells (RBCs) from anemic patients. In the aortic rings from anemic mice, the role of arginases was assessed using either an arginase inhibitor (Nor-NOHA) or genetic ablation of arginase 1 in the endothelium. Inflammatory changes in plasma of CA mice were examined by ELISA. Expression of endothelial NO synthase (eNOS), inducible NO synthase (iNOS), myeloperoxidase (MPO), 3-Nitrotyrosine levels, and 4-Hydroxynonenal (4-HNE) were assessed either by Western blotting or immunohistochemistry. The role of reactive oxygen species (ROS) in ED was assessed in the anemic mice either supplemented with N-Acetyl cysteine (NAC) or by in vitro pharmacological inhibition of MPO. Results The FMD responses were diminished with a correlation to the duration of anemia. Aortic rings from CA mice showed reduced NO-dependent relaxation compared to non-anemic mice. RBCs from anemic patients attenuated NO-dependent relaxation responses in murine aortic rings compared to non-anemic controls. CA results in increased plasma VCAM-1, ICAM-1 levels, and an increased iNOS expression in aortic vascular smooth muscle cells. Arginases inhibition or arginase1 deletion did not improve ED in anemic mice. Increased expression of MPO and 4-HNE observed in endothelial cells of aortic sections from CA mice. NAC supplementation or inhibition of MPO improved relaxation responses in CA mice. Conclusion Chronic anemia is associated with progressive endothelial dysfunction evidenced by activation of the endothelium mediated by systemic inflammation, increased iNOS activity, and ROS production in the arterial wall. ROS scavenger (NAC) supplementation or MPO inhibition are potential therapeutic options to reverse the devastating endothelial dysfunction in chronic anemia.
Collapse
Affiliation(s)
- Ramesh Chennupati
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Isabella Solga
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Patricia Wischmann
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Paul Dahlmann
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Feyza Gül Celik
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Daniela Pacht
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Aslıhan Şahin
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Vithya Yogathasan
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Mohammad Rabiul Hosen
- Department of Internal Medicine II, HeartCenter Bonn, University Hospital Bonn, Bonn, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christian Jung
- Division of Cardiology, Pulmonology and Vascular Medicine, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
- Cardiovascular Research Institute Düsseldorf (CARID), Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
2
|
Janaszak-Jasiecka A, Płoska A, Wierońska JM, Dobrucki LW, Kalinowski L. Endothelial dysfunction due to eNOS uncoupling: molecular mechanisms as potential therapeutic targets. Cell Mol Biol Lett 2023; 28:21. [PMID: 36890458 PMCID: PMC9996905 DOI: 10.1186/s11658-023-00423-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/19/2023] [Indexed: 03/10/2023] Open
Abstract
Nitric oxide (NO) is one of the most important molecules released by endothelial cells, and its antiatherogenic properties support cardiovascular homeostasis. Diminished NO bioavailability is a common hallmark of endothelial dysfunction underlying the pathogenesis of the cardiovascular disease. Vascular NO is synthesized by endothelial nitric oxide synthase (eNOS) from the substrate L-arginine (L-Arg), with tetrahydrobiopterin (BH4) as an essential cofactor. Cardiovascular risk factors such as diabetes, dyslipidemia, hypertension, aging, or smoking increase vascular oxidative stress that strongly affects eNOS activity and leads to eNOS uncoupling. Uncoupled eNOS produces superoxide anion (O2-) instead of NO, thus becoming a source of harmful free radicals exacerbating the oxidative stress further. eNOS uncoupling is thought to be one of the major underlying causes of endothelial dysfunction observed in the pathogenesis of vascular diseases. Here, we discuss the main mechanisms of eNOS uncoupling, including oxidative depletion of the critical eNOS cofactor BH4, deficiency of eNOS substrate L-Arg, or accumulation of its analog asymmetrical dimethylarginine (ADMA), and eNOS S-glutathionylation. Moreover, potential therapeutic approaches that prevent eNOS uncoupling by improving cofactor availability, restoration of L-Arg/ADMA ratio, or modulation of eNOS S-glutathionylation are briefly outlined.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Joanna M Wierońska
- Department of Neurobiology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, 31-343, Kraków, Poland
| | - Lawrence W Dobrucki
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL, 61801, USA.,Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, Urbana, IL, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland. .,BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Gabriela Narutowicza Street, 80-233, Gdansk, Poland.
| |
Collapse
|
3
|
Alzayadneh EM, Shatanawi A, Caldwell RW, Caldwell RB. Methylglyoxal-Modified Albumin Effects on Endothelial Arginase Enzyme and Vascular Function. Cells 2023; 12:795. [PMID: 36899931 PMCID: PMC10001288 DOI: 10.3390/cells12050795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 03/08/2023] Open
Abstract
Advanced glycation end products (AGEs) contribute significantly to vascular dysfunction (VD) in diabetes. Decreased nitric oxide (NO) is a hallmark in VD. In endothelial cells, NO is produced by endothelial NO synthase (eNOS) from L-arginine. Arginase competes with NOS for L-arginine to produce urea and ornithine, limiting NO production. Arginase upregulation was reported in hyperglycemia; however, AGEs' role in arginase regulation is unknown. Here, we investigated the effects of methylglyoxal-modified albumin (MGA) on arginase activity and protein expression in mouse aortic endothelial cells (MAEC) and on vascular function in mice aortas. Exposure of MAEC to MGA increased arginase activity, which was abrogated by MEK/ERK1/2 inhibitor, p38 MAPK inhibitor, and ABH (arginase inhibitor). Immunodetection of arginase revealed MGA-induced protein expression for arginase I. In aortic rings, MGA pretreatment impaired acetylcholine (ACh)-induced vasorelaxation, which was reversed by ABH. Intracellular NO detection by DAF-2DA revealed blunted ACh-induced NO production with MGA treatment that was reversed by ABH. In conclusion, AGEs increase arginase activity probably through the ERK1/2/p38 MAPK pathway due to increased arginase I expression. Furthermore, AGEs impair vascular function that can be reversed by arginase inhibition. Therefore, AGEs may be pivotal in arginase deleterious effects in diabetic VD, providing a novel therapeutic target.
Collapse
Affiliation(s)
- Ebaa M. Alzayadneh
- Department of Physiology and Biochemistry, School of Medicine, University of Jordan, Amman 11942, Jordan
| | - Alia Shatanawi
- Department of Pharmacology, School of Medicine, University of Jordan, Amman 11942, Jordan
| | - R. William Caldwell
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA 30912, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Ruth B. Caldwell
- Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
4
|
Ren Y, Li Z, Li W, Fan X, Han F, Huang Y, Yu Y, Qian L, Xiong Y. Arginase: Biological and Therapeutic Implications in Diabetes Mellitus and Its Complications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2419412. [PMID: 36338341 PMCID: PMC9629921 DOI: 10.1155/2022/2419412] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/18/2022] [Indexed: 09/21/2023]
Abstract
Arginase is a ubiquitous enzyme in the urea cycle (UC) that hydrolyzes L-arginine to urea and L-ornithine. Two mammalian arginase isoforms, arginase1 (ARG1) and arginase2 (ARG2), play a vital role in the regulation of β-cell functions, insulin resistance (IR), and vascular complications via modulating L-arginine metabolism, nitric oxide (NO) production, and inflammatory responses as well as oxidative stress. Basic and clinical studies reveal that abnormal alterations of arginase expression and activity are strongly associated with the onset and development of diabetes mellitus (DM) and its complications. As a result, targeting arginase may be a novel and promising approach for DM treatment. An increasing number of arginase inhibitors, including chemical and natural inhibitors, have been developed and shown to protect against the development of DM and its complications. In this review, we discuss the fundamental features of arginase. Next, the regulatory roles and underlying mechanisms of arginase in the pathogenesis and progression of DM and its complications are explored. Furthermore, we review the development and discuss the challenges of arginase inhibitors in treating DM and its related pathologies.
Collapse
Affiliation(s)
- Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Zhuozhuo Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Wenqing Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Xiaobin Fan
- Department of Obstetrics and Gynecology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China
| | - Feifei Han
- Department of Endocrinology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China
| | - Yaoyao Huang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yi Yu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Department of Obstetrics and Gynecology, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, The Affiliated Hospital of Northwest University, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| |
Collapse
|
5
|
Esposito A, Cotta CK, Lacchini R. Beyond eNOS: Genetic influence in NO pathway affecting drug response. Genet Mol Biol 2022; 45:e20220157. [PMID: 36264109 PMCID: PMC9583294 DOI: 10.1590/1678-4685-gmb-2022-0157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/08/2022] [Indexed: 11/04/2022] Open
Abstract
Nitric Oxide (NO) has important biological functions, and its production may be
influenced by genetic polymorphisms. Since NO mediates the drug response, the
same genetic polymorphism that alter NO levels may also impact drug therapy. The
vast majority of studies in the literature that assess the genetic influence on
NO-related drug response focus on NOS3 (which encodes
endothelial nitric oxide synthase), however several other proteins are
interconnected in the same pathway and may also impact NO availability and drug
response. The aim of this study was to review the literature regarding genetic
polymorphisms that influence NO in response to pharmacological agents located in
genes other than NOS3. Articles were obtained from Pubmed and
consisted of 17 manuscripts that assessed polymorphisms of the following
targets: Arginases 1 and 2 (ARG1 and ARG2),
dimethylarginine dimethylaminohydrolases 1 and 2 (DDAH1 and
DDAH2), and vascular endothelial growth factor
(VEGF). Here we analyze the main results of these articles,
which show promising evidences that may suggest that the NO-driven
pharmacological response is affected by more than the eNOS gene. The search for
genetic markers may result in better understanding of the variability of drug
response and turn pharmacotherapy involving NO safer and more effective.
Collapse
Affiliation(s)
- Aline Esposito
- Universidade de São Paulo, Departamento de Farmacologia, Ribeirão
Preto, São Paulo, SP, Brazil
| | - Cezar Kayzuka Cotta
- Universidade de São Paulo, Departamento de Farmacologia, Ribeirão
Preto, São Paulo, SP, Brazil
| | - Riccardo Lacchini
- Universidade de São Paulo, Departamento de Enfermagem Psiquiátrica e
Ciências Humanas, Ribeirão Preto, São Paulo, SP, Brazil
| |
Collapse
|
6
|
Li Z, Wang L, Ren Y, Huang Y, Liu W, Lv Z, Qian L, Yu Y, Xiong Y. Arginase: shedding light on the mechanisms and opportunities in cardiovascular diseases. Cell Death Dis 2022; 8:413. [PMID: 36209203 PMCID: PMC9547100 DOI: 10.1038/s41420-022-01200-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022]
Abstract
Arginase, a binuclear manganese metalloenzyme in the urea, catalyzes the hydrolysis of L-arginine to urea and L-ornithine. Both isoforms, arginase 1 and arginase 2 perform significant roles in the regulation of cellular functions in cardiovascular system, such as senescence, apoptosis, proliferation, inflammation, and autophagy, via a variety of mechanisms, including regulating L-arginine metabolism and activating multiple signal pathways. Furthermore, abnormal arginase activity contributes to the initiation and progression of a variety of CVDs. Therefore, targeting arginase may be a novel and promising approach for CVDs treatment. In this review, we give a comprehensive overview of the physiological and biological roles of arginase in a variety of CVDs, revealing the underlying mechanisms of arginase mediating vascular and cardiac function, as well as shedding light on the novel and promising therapeutic approaches for CVDs therapy in individuals.
Collapse
Affiliation(s)
- Zhuozhuo Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Liwei Wang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yaoyao Huang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Wenxuan Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Ziwei Lv
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China. .,Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China.
| | - Yi Yu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China. .,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China.
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China. .,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China.
| |
Collapse
|
7
|
Detroja TS, Samson AO. Virtual Screening for FDA-Approved Drugs That Selectively Inhibit Arginase Type 1 and 2. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27165134. [PMID: 36014374 PMCID: PMC9416497 DOI: 10.3390/molecules27165134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 08/05/2022] [Accepted: 08/09/2022] [Indexed: 11/30/2022]
Abstract
Arginases are often overexpressed in human diseases, and they are an important target for developing anti-aging and antineoplastic drugs. Arginase type 1 (ARG1) is a cytosolic enzyme, and arginase type 2 (ARG2) is a mitochondrial one. In this study, a dataset containing 2115-FDA-approved drug molecules is virtually screened for potential arginase binding using molecular docking against several ARG1 and ARG2 structures. The potential arginase ligands are classified into three categories: (1) Non-selective, (2) ARG1 selective, and (3) ARG2 selective. The evaluated potential arginase ligands are then compared with their clinical use. Remarkably, half of the top 30 potential drugs are used clinically to lower blood pressure and treat cancer, infection, kidney disease, and Parkinson’s disease thus partially validating our virtual screen. Most notable are the antihypertensive drugs candesartan, irbesartan, indapamide, and amiloride, the antiemetic rolapitant, the anti-angina ivabradine, and the antidiabetic metformin which have minimal side effects. The partial validation also favors the idea that the other half of the top 30 potential drugs could be used in therapeutic settings. The three categories greatly expand the selectivity of arginase inhibition.
Collapse
|
8
|
Association of cerebral microvascular dysfunction and white matter injury in Alzheimer's disease. GeroScience 2022; 44:1-14. [PMID: 35612774 PMCID: PMC9617002 DOI: 10.1007/s11357-022-00585-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/05/2022] [Indexed: 12/23/2022] Open
Abstract
Patients with Alzheimer's disease (AD) often have cerebral white matter (WM) hyperintensities on MRI and microinfarcts of presumed microvascular origin pathologically. Here, we determined if vasodilator dysfunction of WM-penetrating arterioles is associated with pathologically defined WM injury and disturbances in quantitative MRI-defined WM integrity in patients with mixed microvascular and AD pathology. We analyzed tissues from 28 serially collected human brains from research donors diagnosed with varying degrees of AD neuropathologic change (ADNC) with or without cerebral microinfarcts (mVBI). WM-penetrating and pial surface arteriolar responses to the endothelium-dependent agonist bradykinin were quantified ex vivo with videomicroscopy. Vascular endothelial nitric oxide synthase (eNOS) and NAD(P)H-oxidase (Nox1, 2 and 4 isoforms) expression were measured with quantitative PCR. Glial fibrillary acidic protein (GFAP)-labeled astrocytes were quantified by unbiased stereological approaches in regions adjacent to the sites of WM-penetrating vessel collection. Post-mortem diffusion tensor imaging (DTI) was used to measure mean apparent diffusion coefficient (ADC) and fractional anisotropy (FA), quantitative indices of WM integrity. In contrast to pial surface arterioles, white matter-penetrating arterioles from donors diagnosed with high ADNC and mVBI exhibited a significantly reduced dilation in response to bradykinin when compared to the other groups. Expression of eNOS was reduced, whereas Nox1 expression was increased in WM arterioles in AD and mVBI cases. WM astrocyte density was increased in AD and mVBI, which correlated with a reduced vasodilation in WM arterioles. Moreover, in cases with low ADNC, bradykinin-induced WM arteriole dilation correlated with lower ADC and higher FA values. Comorbid ADNC and mVBI appear to synergistically interact to selectively impair bradykinin-induced vasodilation in WM-penetrating arterioles, which may be related to reduced nitric oxide- and excess reactive oxygen species-mediated vascular endothelial dysfunction. WM arteriole vasodilator dysfunction is associated with WM injury, as supported by reactive astrogliosis and MRI-defined disrupted WM microstructural integrity.
Collapse
|
9
|
Sabe SA, Feng J, Sellke FW, Abid MR. Mechanisms and clinical implications of endothelium-dependent vasomotor dysfunction in coronary microvasculature. Am J Physiol Heart Circ Physiol 2022; 322:H819-H841. [PMID: 35333122 PMCID: PMC9018047 DOI: 10.1152/ajpheart.00603.2021] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 03/21/2022] [Accepted: 03/21/2022] [Indexed: 12/16/2022]
Abstract
Coronary microvascular disease (CMD), which affects the arterioles and capillary endothelium that regulate myocardial perfusion, is an increasingly recognized source of morbidity and mortality, particularly in the setting of metabolic syndrome. The coronary endothelium plays a pivotal role in maintaining homeostasis, though factors such as diabetes, hypertension, hyperlipidemia, and obesity can contribute to endothelial injury and consequently arteriolar vasomotor dysfunction. These disturbances in the coronary microvasculature clinically manifest as diminished coronary flow reserve, which is a known independent risk factor for cardiac death, even in the absence of macrovascular atherosclerotic disease. Therefore, a growing body of literature has examined the molecular mechanisms by which coronary microvascular injury occurs at the level of the endothelium and the consequences on arteriolar vasomotor responses. This review will begin with an overview of normal coronary microvascular physiology, modalities of measuring coronary microvascular function, and clinical implications of CMD. These introductory topics will be followed by a discussion of recent advances in the understanding of the mechanisms by which inflammation, oxidative stress, insulin resistance, hyperlipidemia, hypertension, shear stress, endothelial cell senescence, and tissue ischemia dysregulate coronary endothelial homeostasis and arteriolar vasomotor function.
Collapse
Affiliation(s)
- Sharif A Sabe
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Jun Feng
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - Frank W Sellke
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| | - M Ruhul Abid
- Cardiovascular Research Center, Rhode Island Hospital, Providence, Rhode Island
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, Rhode Island
| |
Collapse
|
10
|
Li X, Zhao W, Peng L, Li Y, Nie S, Yu H, Qin Y, Zhang H. Elevated serum extracellular vesicle arginase 1 in type 2 diabetes mellitus: a cross-sectional study in middle-aged and elderly population. BMC Endocr Disord 2022; 22:62. [PMID: 35277141 PMCID: PMC8917686 DOI: 10.1186/s12902-022-00982-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 03/01/2022] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Serum extracellular vesicle (EV)-derived arginase 1 (ARG 1) plays a critical role in diabetes-associated endothelial dysfunction. This study was performed to determine the levels of serum EV-derived ARG 1 in T2DM and non-T2DM participants and to examine the association of serum EV-derived ARG 1 with T2DM incidence. METHODS We performed a cross-sectional study in 103 Chinese, including 73 T2DM patients and 30 non-T2DM. Serum EVs were prepared via ultracentrifugation. Serum EV-derived ARG 1 levels were measured by enzyme-linked immunosorbent assay. The correlations between serum EV-derived ARG 1 and clinical variables were analyzed. The association of serum EV-derived ARG 1 levels with T2DM was determined by multivariate logistic regression analysis. Interaction subgroup analysis was used to evaluate the interaction of the relevant baselines on the association between serum EV-derived ARG 1 levels and T2DM. RESULTS Serum EV-derived ARG 1 levels were significantly higher in T2DM patients compared with non-T2DM patients (p < 0.001). Correlation analysis revealed that serum EV-derived ARG 1 levels were positively associated with fasting plasma glucose (FPG) (r = 0.316, p = 0.001) and glycated hemoglobin (HbA1c) (r = 0.322, p = 0.001). Serum EV-derived ARG 1 levels were significantly associated with T2DM, especially in the subgroup of T2DM for more than 10 years (OR 1.651, 95% CI = 1.066-2.557; P value, 0.025), after adjusting for confounding factors. CONCLUSIONS Elevated concentration of serum EV-derived ARG 1 is closely associated with T2DM.
Collapse
Affiliation(s)
- Xinwei Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Wen Zhao
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Lu Peng
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Yu Li
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Shaoping Nie
- Department of Emergency, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Huahui Yu
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Yanwen Qin
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China
| | - Huina Zhang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Disease, No. 2 Anzhen Road, Beijing, 100029, China.
| |
Collapse
|
11
|
Bunch KL, Abdelrahman AA, Caldwell RB, Caldwell RW. Novel Therapeutics for Diabetic Retinopathy and Diabetic Macular Edema: A Pathophysiologic Perspective. Front Physiol 2022; 13:831616. [PMID: 35250632 PMCID: PMC8894892 DOI: 10.3389/fphys.2022.831616] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) and diabetic macular edema (DME) are retinal complications of diabetes that can lead to loss of vision and impaired quality of life. The current gold standard therapies for treatment of DR and DME focus on advanced disease, are invasive, expensive, and can trigger adverse side-effects, necessitating the development of more effective, affordable, and accessible therapies that can target early stage disease. The pathogenesis and pathophysiology of DR is complex and multifactorial, involving the interplay between the effects of hyperglycemia, hyperlipidemia, hypoxia, and production of reactive oxygen species (ROS) in the promotion of neurovascular dysfunction and immune cell polarization to a proinflammatory state. The pathophysiology of DR provides several therapeutic targets that have the potential to attenuate disease progression. Current novel DR and DME therapies under investigation include erythropoietin-derived peptides, inducers of antioxidant gene expression, activators of nitric oxide/cyclic GMP signaling pathways, and manipulation of arginase activity. This review aims to aid understanding of DR and DME pathophysiology and explore novel therapies that capitalize on our knowledge of these diabetic retinal complications.
Collapse
Affiliation(s)
- Katharine L. Bunch
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ammar A. Abdelrahman
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Ruth B. Caldwell
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - R. William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA, United States
- James and Jean Culver Vision Discovery Institute, Medical College of Georgia, Augusta University, Augusta, GA, United States
- *Correspondence: R. William Caldwell,
| |
Collapse
|
12
|
Tian Y, Fopiano KA, Patel VS, Feher A, Bagi Z. Role of Caveolae in the Development of Microvascular Dysfunction and Hyperglycemia in Type 2 Diabetes. Front Physiol 2022; 13:825018. [PMID: 35250626 PMCID: PMC8894849 DOI: 10.3389/fphys.2022.825018] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/28/2022] [Indexed: 11/13/2022] Open
Abstract
In type 2 diabetes (T2D) microvascular dysfunction can interfere with tissue glucose uptake thereby contributing to the development of hyperglycemia. The cell membrane caveolae orchestrate signaling pathways that include microvascular control of tissue perfusion. In this study, we examined the role of caveolae in the regulation of microvascular vasomotor function under the condition of hyperglycemia in T2D patients and rodent models. Human coronary arterioles were obtained during cardiac surgery from T2D patients, with higher perioperative glucose levels, and from normoglycemic, non-diabetic controls. The coronary arteriole responses to pharmacological agonists bradykinin and acetylcholine were similar in T2D and non-diabetic patients, however, exposure of the isolated arteries to methyl-β-cyclodextrin (mβCD), an agent known to disrupt caveolae, reduced vasodilation to bradykinin selectively in T2D subjects and converted acetylcholine-induced vasoconstriction to dilation similarly in the two groups. Dilation to the vascular smooth muscle acting nitric oxide donor, sodium nitroprusside, was not affected by mβCD in either group. Moreover, mβCD reduced endothelium-dependent arteriolar dilation to a greater extent in hyperglycemic and obese db/db mice than in the non-diabetic controls. Mechanistically, when fed a high-fat diet (HFD), caveolin-1 knockout mice, lacking caveolae, exhibited a significantly reduced endothelium-dependent arteriolar dilation, both ex vivo and in vivo, which was accompanied by significantly higher serum glucose levels, when compared to HFD fed wild type controls. Thus, in T2D arterioles the role of caveolae in regulating endothelium-dependent arteriole dilation is altered, which appears to maintain vasodilation and mitigate the extent of hyperglycemia. While caveolae play a unique role in microvascular vasomotor regulation, under the condition of hyperglycemia arterioles from T2D subjects appear to be more susceptible for caveolae disruption-associated vasomotor dysfunction and impaired glycemic control.
Collapse
Affiliation(s)
- Yanna Tian
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Katie Anne Fopiano
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Vijay S. Patel
- Department of Surgery, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Attila Feher
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
13
|
A Cross-Talk between the Erythrocyte L-Arginine/ADMA/Nitric Oxide Metabolic Pathway and the Endothelial Function in Subjects with Type 2 Diabetes Mellitus. Nutrients 2021; 13:nu13072306. [PMID: 34371816 PMCID: PMC8308357 DOI: 10.3390/nu13072306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022] Open
Abstract
(1) Background: Type-2-diabetes-mellitus (DM) is one the most important cardiovascular-risk-factors. Among many molecules regulating vascular tone, nitric oxide appears to be the most pivotal. Although micro- and macrovascular-abnormalities are extensively studied, the alterations in the nitric-oxide-metabolic-pathway require further investigations. Additionally, the role of erythrocytes in the vascular tone regulation has not been extensively explored. The aim of this study was to evaluate the endothelial-function and the nitric-oxide-metabolic-pathway in erythrocytes and plasma of diabetic individuals. (2) Methods: A total of 80 subjects were enrolled in this cross-sectional study, including 35 patients with DM and 45 healthy individuals. The endothelial-function was evaluated in response to different stimuli. (3) Results: In the DM group, decreased Arginine and citrulline concentrations in the plasma compartment with reduced Arginine/ADMA and ADMA/DMA-ratios were observed. Preserved nitric-oxide-metabolism in erythrocytes with reduced citrulline level and significantly higher NO-bioavailability were noted. Significant endothelial dysfunction in DM individuals was proved in response to the heat-stimulus. (4) Conclusions: DM patients at an early stage of disease show significant differences in the nitric-oxide-metabolic-pathway, which are more pronounced in the plasma compartment. Erythrocytes constitute a buffer with a higher nitric-oxide-bioavailability, less affected by the DM-related deviations. Patients at an early-stage of DM reveal endothelial-dysfunction, which could be diagnosed earlier using the laser-Doppler-flowmetry.
Collapse
|
14
|
Development of a new nano arginase HPLC capillary column for the fast screening of arginase inhibitors and evaluation of their binding affinity. J Chromatogr B Analyt Technol Biomed Life Sci 2021; 1175:122751. [PMID: 33991957 DOI: 10.1016/j.jchromb.2021.122751] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 11/21/2022]
Abstract
A simple and rapid Nano LC method has been developed for the screening of arginase inhibitors. The method is based on the immobilization of biotinylated arginase on a neutravidin functionalized nano HPLC capillary column. The arginase immobilization step performed by frontal analysis is very fast and only takes a few minutes. The miniaturized capillary column of 170 nL (length 5 cm, internal diameter 75 μm) significantly decreased the required amount of used enzyme (25 pmol). This was of significance importance when working with less available or expensive purified enzyme. Non-selective adsorption of the organic monolith matrix was reduced (<6%) and the arginase efficient yield was high (92%). The resultant affinity capillary columns showed excellent repeatability and long lifetime. The arginase reaction product was achieved within 60 s and the immobilized arginase retained 97% of the initial activity beyond 90 days. This novel approach can thus be used for the fast evaluation of recognition assay induced bya series of inhibitor molecules (caffeic acid phenylamide, chlorogenic acid, piceatannol, nor-NOHA acetate) and plant extracts.
Collapse
|
15
|
Galluccio E, Spadoni S, Fontana B, Bosi E, Piatti P, Monti LD. Long lasting protective effects of early l-arginine treatment on endothelium in an in vitro study. Clin Nutr 2021; 40:1519-1529. [PMID: 33743287 DOI: 10.1016/j.clnu.2021.02.040] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/09/2020] [Accepted: 02/22/2021] [Indexed: 01/11/2023]
Abstract
BACKGROUND & AIMS Excess nutrient supply, such as high fat and high glucose intake, promotes oxidative stress and advanced glycation end products accumulation. Oxidative stress and AGE accumulation cause pathological elevation of arginase activity and pro-inflammatory signaling implicated in endothelial dysfunction. Several studies showed positive effects of l-arginine supplementation in endothelial function but little is currently known about the role of l-arginine as prevention of endothelial dysfunction caused by excessive nutrient supply (overfeeding). Our aim was to evaluate a possible protective effect of l-arginine on endothelial dysfunction caused by excessive nutrient supply (overfeeding), using human endothelial cells line in an in vitro study. METHODS Endothelial EA.hy926 cells were pre-treated with 1.72 mM of l-arginine for 24 h and afterwards subjected to nutritional stress (high lipid, high insulin and high glucose concentrations) for further 24 h. After treatment discontinuation, the cells were kept in culture for 48 h, in physiological condition, to evaluate the effects of treatments after normalization. RESULTS Excess nutrient supply in EA.hy926 cell line showed an increase of oxidative and nitrosative stress, a rise of AGEs production, high arginase activity, leading the cells to acidosis and to cell death. l-arginine pretreatment protects the cells by reducing apoptosis, acidosis, oxidative and nitrosative stress, arginase activity and AGE accumulation. l-arginine pretreatment reduces AGEs generation and accumulation by regulating STAB1 and RAGE gene expression levels. STAB1, acting as receptor scavenger of AGEs, interferes with AGE-RAGE binding and thus prevents activation of intracellular signaling pathways leading to cell damage. Moreover the reduction of oxidative stress promotes a decrease of excessive activation of arginase involved in endothelial dysfunction. The effects of pretreatment with l-arginine last even in the absence of stimuli and despite after treatment discontinuation. CONCLUSIONS An early l-arginine treatment is able to prevent oxidative stress and AGEs accumulation caused by overfeeding in human endothelial cell line by regulating STAB1/RAGE gene expression and by reducing excess arginase activity. The positive effects of l-arginine pretreatment continue even after treatment discontinuation in normal conditions.
Collapse
Affiliation(s)
- Elena Galluccio
- Cardio-Diabetes and Core Lab Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy; Cardio-Metabolism and Clinical Trials Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Serena Spadoni
- Cardio-Diabetes and Core Lab Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy; Cardio-Metabolism and Clinical Trials Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Barbara Fontana
- Cardio-Diabetes and Core Lab Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy; Cardio-Metabolism and Clinical Trials Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Emanuele Bosi
- Cardio-Diabetes and Core Lab Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy; Cardio-Metabolism and Clinical Trials Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Piermarco Piatti
- Cardio-Diabetes and Core Lab Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy; Cardio-Metabolism and Clinical Trials Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy.
| | - Lucilla D Monti
- Cardio-Diabetes and Core Lab Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy; Cardio-Metabolism and Clinical Trials Unit, Diabetes Research Institute, Department of Internal Medicine, IRCCS San Raffaele Institute, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
16
|
Bahadoran Z, Mirmiran P, Kashfi K, Ghasemi A. Endogenous flux of nitric oxide: Citrulline is preferred to Arginine. Acta Physiol (Oxf) 2021; 231:e13572. [PMID: 33089645 DOI: 10.1111/apha.13572] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 09/09/2020] [Accepted: 10/17/2020] [Indexed: 02/06/2023]
Abstract
Both arginine (Arg) and its precursor citrulline (Cit) have received much interest in the past two decades because of their potential effects on whole-body nitric oxide (NO) production and augmentation of NO-dependent signalling pathways. However, the usefulness of Arg supplementation for NO production is questionable because of its high splanchnic first pass metabolism (FPM), which limits its systemic availability. Both hepatic- and extrahepatic arginases critically limit the availability of Arg for the NO synthase enzymes (NOSs) and therefore, a limited amount of oral Arg can reach the systemic circulation for NO synthesis. Arg also has some undesired effects including induction of arginase activity, an increase of urea levels, a decrease of cellular uptake of Cit and decrease of recycling of Arg from Cit. In contrast, Cit has more availability as an NO precursor because of its high intestinal absorption, low FPM and high renal reabsorption. At the cellular level, co-localization of Cit transport systems and the enzymes involved in the Cit-Arg-NO pathway facilitates channelling of Cit into NO. Furthermore, cells preferably use Cit rather than either intra- or extracellular Arg to improve NO output, especially in high-demand situations. In conclusion, available evidence strongly supports the concept that Cit leads to higher NO production and suggests that Cit may have a better therapeutic effect than Arg for NO-disrupted conditions.
Collapse
Affiliation(s)
- Zahra Bahadoran
- Nutrition and Endocrine Research Center Research Institute for Endocrine SciencesShahid Beheshti University of Medical Sciences Tehran Iran
| | - Parvin Mirmiran
- Department of Clinical Nutrition and Human Dietetics Faculty of Nutrition Sciences and Food Technology National Nutrition and Food Technology Research InstituteShahid Beheshti University of Medical Sciences Tehran Iran
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences Sophie Davis School of Biomedical Education City University of New York School of Medicine New York NY USA
| | - Asghar Ghasemi
- Endocrine Physiology Research Center Research Institute for Endocrine SciencesShahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
17
|
Moretto J, Pudlo M, Demougeot C. Human-based evidence for the therapeutic potential of arginase inhibitors in cardiovascular diseases. Drug Discov Today 2020; 26:138-147. [PMID: 33197620 DOI: 10.1016/j.drudis.2020.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/22/2020] [Accepted: 11/05/2020] [Indexed: 01/25/2023]
Affiliation(s)
- Johnny Moretto
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France.
| | - Marc Pudlo
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Céline Demougeot
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
18
|
Endothelial Dysfunction: A Contributor to Adverse Cardiovascular Remodeling and Heart Failure Development in Type 2 Diabetes beyond Accelerated Atherogenesis. J Clin Med 2020; 9:jcm9072090. [PMID: 32635218 PMCID: PMC7408687 DOI: 10.3390/jcm9072090] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction, associated with depressed nitric oxide (NO) bioavailability, is awell-recognized contributor to both accelerated atherogenesis and microvascular complications intype 2 diabetes (DM). However, growing evidence points to the comorbidities-driven endothelialdysfunction within coronary microvessels as a key player responsible for left ventricular (LV)diastolic dysfunction, restrictive LV remodeling and heart failure with preserved ejection fraction(HFpEF), the most common form of heart failure in DM. In this review we have described: (1)multiple cellular pathways which may link depressed NO bioavailability to LV diastolicdysfunction and hypertrophy; (2) hemodynamic consequences and prognostic effects of restrictiveLV remodeling and combined diastolic and mild systolic LV dysfunction on cardiovascularoutcomes in DM and HFpEF, with a focus on the clinical relevance of endothelial dysfunction; (3)novel therapeutic strategies to improve endothelial function in DM. In summary, beyondassociations with accelerated atherogenesis and microvascular complications, endothelialdysfunction supplements the multiple interwoven pathways affecting cardiomyocytes, endothelialcells and the extracellular matrix with consequent LV dysfunction in DM patients. The associationamongst impaired endothelial function, reduced coronary flow reserve, combined LV diastolic anddiscrete systolic dysfunction, and low LV stroke volume and preload reserve-all of which areadverse outcome predictors-is a dangerous constellation of inter-related abnormalities, underlyingthe development of heart failure. Nevertheless, the relevance of endothelial effects of novel drugsin terms of their ability to attenuate cardiovascular remodeling and delay heart failure onset in DMpatients remains to be investigated.
Collapse
|
19
|
Cheng H, Lu T, Wang J, Xia Y, Chai X, Zhang M, Yao Y, Zhou N, Zhou S, Chen X, Su W, Liu C, Yi W, Chen Y, Yao L. HuangqiGuizhiWuwu Decoction Prevents Vascular Dysfunction in Diabetes via Inhibition of Endothelial Arginase 1. Front Physiol 2020; 11:201. [PMID: 32269530 PMCID: PMC7109290 DOI: 10.3389/fphys.2020.00201] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 02/21/2020] [Indexed: 12/12/2022] Open
Abstract
Hyperglycemia induces vascular endothelial dysfunction, which contributes to the development of vascular complication of diabetes. A classic prescription of traditional medicine, HuangqiGuizhiWuwu Decoction (HGWWD) has been used for the treatment of various cardiovascular and cerebrovascular diseases, which all are related with vascular pathology. The present study investigated the effect of HGWWD treatment in streptozocin (STZ)-induced vascular dysfunction in mouse models. In vivo studies were performed using wild type mice as well as arginase 1 knockout specific in endothelial cells (EC-A1-/-) of control mice, diabetes mice and diabetes mice treated with HGWWD (60 g crude drugs/kg/d) for 2 weeks. For in vitro studies, aortic tissues were treated with mice serum containing HGWWD with or without adenoviral arginase 1 (Ad-A1) transduction in high glucose (HG) medium. We found that HGWWD treatment restored STZ-induced impaired mean velocity and pulsatility index of mouse left femoral arteries, aortic pulse wave velocity and vascular endothelial relaxation accompanied by elevated NO production in the aorta and plasma, as well as reduced endothelial arginase activity and aortic arginase 1 expression. The protective effect of HGWWD is reversed by an inhibitor of nitric oxide synthesis. Meanwhile, the preventive effect of serum containing HGWWD in endothelial vascular dysfunction is completely blocked by Ad-A1 transduction in HG incubated aortas. HGWWD treatment further improved endothelial vascular dysfunction in STZ induced EC-A1-/- mice. This study demonstrates that HGWWD improved STZ-induced vascular dysfunction through arginase 1 - NO signaling, specifically targeting endothelial arginase 1.
Collapse
Affiliation(s)
- Hong Cheng
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tian Lu
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jingya Wang
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yucen Xia
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoshu Chai
- Department of Oncology, Guangdong Provincial Hospital of Traditional Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Minyi Zhang
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yutong Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Na Zhou
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sisi Zhou
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xinyi Chen
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weiwei Su
- Guangdong Key Laboratory of Plant Resources, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Cunzhi Liu
- Acupuncture Research Center, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Yi
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongjun Chen
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lin Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Is the Arginase Pathway a Novel Therapeutic Avenue for Diabetic Retinopathy? J Clin Med 2020; 9:jcm9020425. [PMID: 32033258 PMCID: PMC7073619 DOI: 10.3390/jcm9020425] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 01/31/2020] [Indexed: 12/12/2022] Open
Abstract
Diabetic retinopathy (DR) is the leading cause of blindness in working age Americans. Clinicians diagnose DR based on its characteristic vascular pathology, which is evident upon clinical exam. However, extensive research has shown that diabetes causes significant neurovascular dysfunction prior to the development of clinically apparent vascular damage. While laser photocoagulation and/or anti-vascular endothelial growth factor (VEGF) therapies are often effective for limiting the late-stage vascular pathology, we still do not have an effective treatment to limit the neurovascular dysfunction or promote repair during the early stages of DR. This review addresses the role of arginase as a mediator of retinal neurovascular injury and therapeutic target for early stage DR. Arginase is the ureohydrolase enzyme that catalyzes the production of L-ornithine and urea from L-arginine. Arginase upregulation has been associated with inflammation, oxidative stress, and peripheral vascular dysfunction in models of both types of diabetes. The arginase enzyme has been identified as a therapeutic target in cardiovascular disease and central nervous system disease including stroke and ischemic retinopathies. Here, we discuss and review the literature on arginase-induced retinal neurovascular dysfunction in models of DR. We also speculate on the therapeutic potential of arginase in DR and its related underlying mechanisms.
Collapse
|
21
|
Azul L, Leandro A, Boroumand P, Klip A, Seiça R, Sena CM. Increased inflammation, oxidative stress and a reduction in antioxidant defense enzymes in perivascular adipose tissue contribute to vascular dysfunction in type 2 diabetes. Free Radic Biol Med 2020; 146:264-274. [PMID: 31698080 DOI: 10.1016/j.freeradbiomed.2019.11.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 10/19/2019] [Accepted: 11/03/2019] [Indexed: 01/27/2023]
Abstract
BACKGROUND Perivascular adipose tissue (PVAT) surrounds most large blood vessels and plays an important role in vascular homeostasis. The present study was conducted to investigate the contribution of PVAT to vascular dysfunction in a rat model of type 2 diabetes. MATERIAL AND METHODS Several in vivo parameters such as lipid profile (total cholesterol and triglyceride systemic levels), fasting glucose levels, glucose tolerance and insulin sensitivity (through glucose and insulin tolerance tests, respectively) were determined in Goto-Kakizaki (GK) diabetic rats and compared with control Wistar rats. At the vascular level, endothelial dependent and independent relaxation and contraction studies were performed in aortic rings in the absence (PVAT-) or in the presence (PVAT+) of thoracic PVAT. We also evaluated vascular oxidative stress and performed western blots, PCR and immunohistochemistry analysis of cytokines and various enzymes in PVAT. RESULTS Endothelium-dependent relaxation to acetylcholine, assessed by wire myography, was impaired in GK rats and improved by the antioxidant TEMPOL and by the TLR4 inhibitor, CLI-095 suggesting an increase in oxidative stress and inflammation. In addition, vascular superoxide and peroxynitrite production was increased in the vascular wall of diabetic rats, accompanied by reduced nitric oxide bioavailability. The presence of PVAT had an anticontractile effect in response to phenylephrine in Wistar rats that was lost in GK rats. Western blot and immunohistochemistry analysis revealed that PVAT phenotype shifts, under diabetic conditions, towards a proinflammatory (with increment in CRP, CCL2, CD36), pro-oxidant (increased levels of aldose reductase, and reduced levels of antioxidant deference enzymes) and vasoconstriction state. CONCLUSION Our data suggest that this rat model of type 2 diabetes is associated with perivascular adipose dysfunction that contributes to oxidative stress, inflammation and endothelial dysfunction.
Collapse
Affiliation(s)
- Lara Azul
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal
| | - Adriana Leandro
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal
| | - Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, M5G 0A4, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, M5S 1A8, Canada
| | - Raquel Seiça
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal
| | - Cristina M Sena
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Portugal.
| |
Collapse
|
22
|
Mahdi A, Kövamees O, Pernow J. Improvement in endothelial function in cardiovascular disease - Is arginase the target? Int J Cardiol 2019; 301:207-214. [PMID: 31785959 DOI: 10.1016/j.ijcard.2019.11.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/26/2019] [Accepted: 11/04/2019] [Indexed: 01/30/2023]
Abstract
Endothelial dysfunction represents an early change in the vascular wall in areas prone to atherosclerotic plaque formation and is present in association with several risk factors for cardiovascular disease. The underlying mechanisms behind endothelial dysfunction are multifactorial and complex. Arginase has emerged as a key player in the regulation of endothelial integrity by the ability of reciprocally inhibits nitric oxide formation and promoting oxidative stress. A chain of evidence suggest that arginase is implicated in the pathogenesis underlying endothelial dysfunction induced by several cardiovascular risk factors and established cardiovascular disease including diabetes, hypercholesteremia, ischemia/reperfusion, atherosclerosis, obesity, ageing and hypertension. Recent data has unveiled a key role of arginase as one of the key mechanisms underlying endothelial dysfunction in diabetes and may serve as a potential therapeutic target in previously overlooked compartments including red blood cells. The current review is devoted to discuss arginase as a key mediator in endothelial dysfunction and the potential for therapeutic possibilities to target this enzyme in various diseases, especially type 2 diabetes, atherosclerosis and ischemia/reperfusion with focus on translational and clinical aspects. Moreover, approaches of how and in which patient group(s) arginase may be targeted in future clinical trials are discussed.
Collapse
Affiliation(s)
- Ali Mahdi
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden
| | - Oskar Kövamees
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine, Division of Cardiology, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Division, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
23
|
Mazrouei S, Sharifpanah F, Caldwell RW, Franz M, Shatanawi A, Muessig J, Fritzenwanger M, Schulze PC, Jung C. Regulation of MAP kinase-mediated endothelial dysfunction in hyperglycemia via arginase I and eNOS dysregulation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1866:1398-1411. [DOI: 10.1016/j.bbamcr.2019.05.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 12/14/2018] [Accepted: 05/20/2019] [Indexed: 12/24/2022]
|
24
|
Zhou Z, Mahdi A, Tratsiakovich Y, Zahorán S, Kövamees O, Nordin F, Uribe Gonzalez AE, Alvarsson M, Östenson CG, Andersson DC, Hedin U, Hermesz E, Lundberg JO, Yang J, Pernow J. Erythrocytes From Patients With Type 2 Diabetes Induce Endothelial Dysfunction Via Arginase I. J Am Coll Cardiol 2019; 72:769-780. [PMID: 30092954 DOI: 10.1016/j.jacc.2018.05.052] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/05/2018] [Accepted: 05/20/2018] [Indexed: 02/01/2023]
Abstract
BACKGROUND Cardiovascular complications are major clinical problems in type 2 diabetes mellitus (T2DM). The authors previously demonstrated a crucial role of red blood cells (RBCs) in control of cardiac function through arginase-dependent regulation of nitric oxide export from RBCs. There is alteration of RBC function, as well as an increase in arginase activity, in T2DM. OBJECTIVES The authors hypothesized that RBCs from patients with T2DM induce endothelial dysfunction by up-regulation of arginase. METHODS RBCs were isolated from patients with T2DM and age-matched healthy subjects and were incubated with rat aortas or human internal mammary arteries from nondiabetic patients for vascular reactivity and biochemical studies. RESULTS Arginase activity and arginase I protein expression were elevated in RBCs from patients with T2DM (T2DM RBCs) through an effect induced by reactive oxygen species (ROS). Co-incubation of arterial segments with T2DM RBCs, but not RBCs from age-matched healthy subjects, significantly impaired endothelial function but not smooth muscle cell function in both healthy rat aortas and human internal mammary arteries. Endothelial dysfunction induced by T2DM RBCs was prevented by inhibition of arginase and ROS both at the RBC and vascular levels. T2DM RBCs induced increased vascular arginase I expression and activity through an ROS-dependent mechanism. CONCLUSIONS This study demonstrates a novel mechanism behind endothelial dysfunction in T2DM that is induced by RBC arginase I and ROS. Targeting arginase I in RBCs may serve as a novel therapeutic tool for the treatment of endothelial dysfunction in T2DM.
Collapse
Affiliation(s)
- Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Ali Mahdi
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Yahor Tratsiakovich
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Szabolcs Zahorán
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Oskar Kövamees
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Filip Nordin
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | | | - Michael Alvarsson
- Division of Endocrinology and Diabetology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Claes-Göran Östenson
- Division of Endocrinology and Diabetology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Daniel C Andersson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Theme, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Ulf Hedin
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Edit Hermesz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jiangning Yang
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden; Heart and Vascular Theme, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Hein TW, Xu X, Ren Y, Xu W, Tsai SH, Thengchaisri N, Kuo L. Requisite roles of LOX-1, JNK, and arginase in diabetes-induced endothelial vasodilator dysfunction of porcine coronary arterioles. J Mol Cell Cardiol 2019; 131:82-90. [PMID: 31015037 DOI: 10.1016/j.yjmcc.2019.04.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/12/2019] [Accepted: 04/16/2019] [Indexed: 01/05/2023]
Abstract
Diabetes is associated with cardiac inflammation and impaired endothelium-dependent coronary vasodilation, but molecular mechanisms involved in this dysfunction remain unclear. We examined contributions of inflammatory molecules lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1), stress-activated kinases (c-Jun N-terminal kinase [JNK] and p38), arginase, and reactive oxygen species to coronary arteriolar dysfunction in a porcine model of type 1 diabetes. Coronary arterioles were isolated from streptozocin-induced diabetic pigs and control pigs for vasoreactivity and molecular/biochemical studies. Endothelium-dependent nitric oxide (NO)-mediated vasodilation to serotonin was diminished after 2 weeks of diabetes, without altering endothelium-independent vasodilation to sodium nitroprusside. Superoxide scavenger TEMPOL, NO precursor L-arginine, arginase inhibitor nor-NOHA, anti-LOX-1 antibody or JNK inhibitors SP600125 and BI-78D3 improved dilation of diabetic vessels to serotonin. However, hydrogen peroxide scavenger catalase, anti-IgG antibody or p38 kinase inhibitor SB203580 had no effect. Combined inhibition of arginase and superoxide levels did not further improve vasodilation. Arginase-I mRNA expression, LOX-1 and JNK protein expression, and superoxide levels were elevated in diabetic arterioles. In conclusion, sequential activation of LOX-1, JNK, and L-arginine consuming enzyme arginase-I in diabetes elicits superoxide-dependent oxidative stress and impairs endothelial NO-mediated dilation in coronary arterioles. Therapeutic targeting of these adverse vascular molecules may improve coronary arteriolar function during diabetes.
Collapse
Affiliation(s)
- Travis W Hein
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, United States.
| | - Xin Xu
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, United States
| | - Yi Ren
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, United States
| | - Wenjuan Xu
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, United States
| | - Shu-Huai Tsai
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, United States
| | - Naris Thengchaisri
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, United States
| | - Lih Kuo
- Department of Medical Physiology, College of Medicine, Texas A&M University Health Science Center, Temple, TX, United States
| |
Collapse
|
26
|
Moretto J, Girard C, Demougeot C. The role of arginase in aging: A systematic review. Exp Gerontol 2019; 116:54-73. [DOI: 10.1016/j.exger.2018.12.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 12/07/2018] [Accepted: 12/12/2018] [Indexed: 12/15/2022]
|
27
|
Fawad Ali Shah S, Iqbal T, Naveed N, Akram S, Arshad Rafiq M, Hussain S. ARG1 single nucleotide polymorphisms rs2781666 and rs2781665 confer risk of Type 2 diabetes mellitus. EXCLI JOURNAL 2018; 17:847-855. [PMID: 30233283 PMCID: PMC6141821 DOI: 10.17179/excli2018-1178] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 08/20/2018] [Indexed: 11/25/2022]
Abstract
Genetic polymorphisms mapped in the ARG1 locus (chr6:131894344-131905472) and their functional effects on type 2 diabetes mellitus (T2DM) have not been thoroughly elucidated to date. The present study aimed to investigate an association between variant alleles at ARG1 locus and T2DM in patients. Two ARG1 single nucleotide polymorphisms (SNPs) were characterized in a representative sample of 500 patients with T2DM and 500 healthy volunteers. Serum lipid profile was studied by spectrophotometric analysis, while serum arginase-1 concentrations were determined by an enzyme-linked immunosorbent assay. The regions, encompassing target SNPs (rs2781665 and rs2781666), were amplified by polymerase chain reaction and genotypes were assigned by restriction digestions. A statistically significant increase was observed in the serum hs-CRP and arginase-1 levels in the subjects with T2DM than in controls (P <0.0001; for each). The variant genotypes of rs2781666 and rs2781665 were significantly associated with T2DM when compared with controls (P< 0.0001). Moreover, type 2 diabetic patients showed higher frequencies of T allele at rs2781666 and rs2781665 compared to the controls (OR = 1.7; 95 % CI=1.31-2.13; P <0.0001, and OR = 1.9; 95 % CI=1.45-2.38; P <0.0001, respectively). Haplotype T-T (chr6: 131893247-131893559) mapped at rs2781665-A/T and rs2781666-G/T displays higher frequency in the subjects when compared to the healthy ethnically-matched control samples (P <0.0001). We wish to propose, the first ever observation to our knowledge that concluding high levels of arginase-1 and the ARG1 polymorphisms are possible causes to confer/augment the risk of T2DM in subjects originates in Pakistan.
Collapse
Affiliation(s)
- Syed Fawad Ali Shah
- Department of Biosciences, COMSATS University Islamabad, Park Road, Chak Shehzad, Islamabad 45550, Pakistan
| | - Tahir Iqbal
- Department of Internal Medicine, Shifa College of Medicine, Shifa International Hospital, H-8/4, Islamabad 44000, Pakistan
| | - Nasreen Naveed
- The Diabetic Centre, Phulgran Stop Near Toll Plaza, Murree Express Way, Islamabad 635, Pakistan
| | - Sumaira Akram
- Department of Biosciences, COMSATS University Islamabad, Park Road, Chak Shehzad, Islamabad 45550, Pakistan
| | - Muhammad Arshad Rafiq
- Department of Biosciences, COMSATS University Islamabad, Park Road, Chak Shehzad, Islamabad 45550, Pakistan
| | - Sabir Hussain
- Department of Biosciences, COMSATS University Islamabad, Park Road, Chak Shehzad, Islamabad 45550, Pakistan
| |
Collapse
|
28
|
Serum exosomes mediate delivery of arginase 1 as a novel mechanism for endothelial dysfunction in diabetes. Proc Natl Acad Sci U S A 2018; 115:E6927-E6936. [PMID: 29967177 DOI: 10.1073/pnas.1721521115] [Citation(s) in RCA: 90] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Exosomes, abundant in blood, deliver various molecules to recipient cells. Endothelial cells are directly exposed to circulating substances. However, how endothelial cells respond to serum exosomes (SExos) and the implications in diabetes-associated vasculopathy have never been explored. In the present study, we showed that SExos from diabetic db/db mice (db/db SExos) were taken up by aortic endothelial cells, which severely impaired endothelial function in nondiabetic db/m+ mice. The exosomal proteins, rather than RNAs, mostly account for db/db SExos-induced endothelial dysfunction. Comparative proteomics analysis showed significant increase of arginase 1 in db/db SExos. Silence or overexpression of arginase 1 confirmed its essential role in db/db SExos-induced endothelial dysfunction. This study is a demonstration that SExos deliver arginase 1 protein to endothelial cells, representing a cellular mechanism during development of diabetic endothelial dysfunction. The results expand the scope of blood-borne substances that monitor vascular homeostasis.
Collapse
|
29
|
Chennupati R, Meens MJ, Janssen BJ, van Dijk P, Hakvoort TBM, Lamers WH, De Mey JGR, Koehler SE. Deletion of endothelial arginase 1 does not improve vasomotor function in diabetic mice. Physiol Rep 2018; 6:e13717. [PMID: 29890043 PMCID: PMC5995309 DOI: 10.14814/phy2.13717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 01/08/2023] Open
Abstract
Endothelial arginase 1 was ablated to assess whether this prevents hyperglycemia-induced endothelial dysfunction by improving arginine availability for nitric oxide production. Endothelial Arg1-deficient mice (Arg1-KOTie2 ) were generated by crossing Arg1fl/fl (controls) with Tie2Cretg/- mice and analyzed by immunohistochemistry, measurements of hemodynamics, and wire myography. Ablation was confirmed by immunohistochemistry. Mean arterial blood pressure was similar in conscious male control and Arg1-KOTie2 mice. Depletion of circulating arginine by intravenous infusion of arginase 1 or inhibition of nitric oxide synthase activity with L-NG -nitro-arginine methyl ester increased mean arterial pressure similarly in control (9 ± 2 and 34 ± 2 mmHg, respectively) and Arg1-KOTie2 mice (11 ± 3 and 38 ± 4 mmHg, respectively). Vasomotor responses were studied in isolated saphenous arteries of 12- and 34-week-old Arg1-KOTie2 and control animals by wire myography. Diabetes was induced in 10-week-old control and Arg1-KOTie2 mice with streptozotocin, and vasomotor responses were studied 10 weeks later. Optimal arterial diameter, contractile responses to phenylephrine, and relaxing responses to acetylcholine and sodium nitroprusside were similar in normoglycemic control and Arg1-KOTie2 mice. The relaxing response to acetylcholine was dependent on the availability of extracellular l-arginine. In the diabetic mice, arterial relaxation responses to endothelium-dependent hyperpolarization and to exogenous nitric oxide were impaired. The data show that endothelial ablation of arginase 1 in mice does not markedly modify smooth muscle and endothelial functions of a resistance artery under normo- and hyperglycemic conditions.
Collapse
Affiliation(s)
- Ramesh Chennupati
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtthe Netherlands
- Nutrim ‐ School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| | - Merlijn J. Meens
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Department of Pathology and ImmunologyUniversity of GenevaGenevaSwitzerland
| | - Ben J. Janssen
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtthe Netherlands
| | - Paul van Dijk
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
| | | | - Wouter H. Lamers
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
- Nutrim ‐ School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| | - Jo G. R. De Mey
- Department of Pharmacology & ToxicologyMaastricht UniversityMaastrichtthe Netherlands
- Cardiovascular Research Institute Maastricht (CARIM)Maastricht UniversityMaastrichtthe Netherlands
- Department of Cardiovascular and Renal ResearchInstitute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of CardiacThoracic and Vascular SurgeryOdense University HospitalOdenseDenmark
| | - S. Eleonore Koehler
- Departments of Anatomy & EmbryologyMaastricht UniversityMaastrichtthe Netherlands
- Nutrim ‐ School of Nutrition and Translational Research in MetabolismMaastricht UniversityMaastrichtthe Netherlands
| |
Collapse
|
30
|
Shosha E, Xu Z, Narayanan SP, Lemtalsi T, Fouda AY, Rojas M, Xing J, Fulton D, Caldwell RW, Caldwell RB. Mechanisms of Diabetes-Induced Endothelial Cell Senescence: Role of Arginase 1. Int J Mol Sci 2018; 19:ijms19041215. [PMID: 29673160 PMCID: PMC5979610 DOI: 10.3390/ijms19041215] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 04/13/2018] [Accepted: 04/14/2018] [Indexed: 12/17/2022] Open
Abstract
We have recently found that diabetes-induced premature senescence of retinal endothelial cells is accompanied by NOX2-NADPH oxidase-induced increases in the ureohydrolase enzyme arginase 1 (A1). Here, we used genetic strategies to determine the specific involvement of A1 in diabetes-induced endothelial cell senescence. We used A1 knockout mice and wild type mice that were rendered diabetic with streptozotocin and retinal endothelial cells (ECs) exposed to high glucose or transduced with adenovirus to overexpress A1 for these experiments. ABH [2(S)-Amino-6-boronohexanoic acid] was used to inhibit arginase activity. We used Western blotting, immunolabeling, quantitative PCR, and senescence associated β-galactosidase (SA β-Gal) activity to evaluate senescence. Analyses of retinal tissue extracts from diabetic mice showed significant increases in mRNA expression of the senescence-related proteins p16INK4a, p21, and p53 when compared with non-diabetic mice. SA β-Gal activity and p16INK4a immunoreactivity were also increased in retinal vessels from diabetic mice. A1 gene deletion or pharmacological inhibition protected against the induction of premature senescence. A1 overexpression or high glucose treatment increased SA β-Gal activity in cultured ECs. These results demonstrate that A1 is critically involved in diabetes-induced senescence of retinal ECs. Inhibition of arginase activity may therefore be an effective therapeutic strategy to alleviate diabetic retinopathy by preventing premature senescence.
Collapse
Affiliation(s)
- Esraa Shosha
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Zhimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - S Priya Narayanan
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
- Department of Occupational Therapy, College of Allied Health Sciences, Augusta University, Augusta, GA 30912, USA.
| | - Tahira Lemtalsi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Abdelrahman Y Fouda
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Modesto Rojas
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| | - Ji Xing
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - David Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA 30904, USA.
| |
Collapse
|
31
|
Caldwell RW, Rodriguez PC, Toque HA, Narayanan SP, Caldwell RB. Arginase: A Multifaceted Enzyme Important in Health and Disease. Physiol Rev 2018; 98:641-665. [PMID: 29412048 PMCID: PMC5966718 DOI: 10.1152/physrev.00037.2016] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 08/14/2017] [Accepted: 08/17/2017] [Indexed: 12/15/2022] Open
Abstract
The arginase enzyme developed in early life forms and was maintained during evolution. As the last step in the urea cycle, arginase cleaves l-arginine to form urea and l-ornithine. The urea cycle provides protection against excess ammonia, while l-ornithine is needed for cell proliferation, collagen formation, and other physiological functions. In mammals, increases in arginase activity have been linked to dysfunction and pathologies of the cardiovascular system, kidney, and central nervous system and also to dysfunction of the immune system and cancer. Two important aspects of the excessive activity of arginase may be involved in diseases. First, overly active arginase can reduce the supply of l-arginine needed for the production of nitric oxide (NO) by NO synthase. Second, too much l-ornithine can lead to structural problems in the vasculature, neuronal toxicity, and abnormal growth of tumor cells. Seminal studies have demonstrated that increased formation of reactive oxygen species and key inflammatory mediators promote this pathological elevation of arginase activity. Here, we review the involvement of arginase in diseases affecting the cardiovascular, renal, and central nervous system and cancer and discuss the value of therapies targeting the elevated activity of arginase.
Collapse
Affiliation(s)
- R William Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Paulo C Rodriguez
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Haroldo A Toque
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - S Priya Narayanan
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| | - Ruth B Caldwell
- Department of Pharmacology & Toxicology, Vision Discovery Institute, Department of Medicine-Hematology and Oncology, Department of Occupational Therapy, School of Allied Health Sciences, and Vascular Biology Center, Medical College of Georgia, Augusta University , Augusta, Georgia ; and VA Medical Center, Augusta, Georgia
| |
Collapse
|
32
|
Differential expression of genes identified by suppression subtractive hybridization in liver and adipose tissue of gerbils with diabetes. PLoS One 2018; 13:e0191212. [PMID: 29394254 PMCID: PMC5796689 DOI: 10.1371/journal.pone.0191212] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 12/29/2017] [Indexed: 11/19/2022] Open
Abstract
Objectives We aimed at identifying genes related to hereditary type 2 diabetes expressed in the liver and the adipose tissue of spontaneous diabetic gerbils using suppression subtractive hybridization (SSH) screening. Methods Two gerbil littermates, one with high and the other with normal blood glucose level, from our previously bred spontaneous diabetic gerbil strain were used in this study. To identify differentially expressed genes in the liver and the adipose tissue, mRNA from these tissues was extracted and SSH libraries were constructed for screening. After sequencing and BLAST analyzing, up or down-regulated genes possibly involved in metabolism and diabetes were selected, and their expression levels in diabetic gerbils and normal controls were analyzed using quantitative RT-PCR and Western blotting. Results A total of 4 SSH libraries were prepared from the liver and the adipose tissue of gerbils. There are 95 up or down-regulated genes were identified to be involved in metabolism, oxidoreduction, RNA binding, cell proliferation, and differentiation or other function. Expression of 17 genes most possibly associated with diabetes was analyzed and seven genes (Sardh, Slc39a7, Pfn1, Arg1, Cth, Sod1 and P4hb) in the liver and one gene (Fabp4) in the adipose tissue were identified that were significantly differentially expressed between diabetic gerbils and control animals. Conclusions We identified eight genes associated with type 2 diabetes from the liver and the adipose tissue of gerbils via SSH screening. These findings provide further insights into the molecular mechanisms of diabetes and imply the value of our spontaneous diabetic gerbil strain as a diabetes model.
Collapse
|
33
|
Bagi Z, Brandner DD, Le P, McNeal DW, Gong X, Dou H, Fulton DJ, Beller A, Ngyuen T, Larson EB, Montine TJ, Keene CD, Back SA. Vasodilator dysfunction and oligodendrocyte dysmaturation in aging white matter. Ann Neurol 2018; 83:142-152. [PMID: 29283444 PMCID: PMC5876126 DOI: 10.1002/ana.25129] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 12/24/2017] [Accepted: 12/26/2017] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Microvascular brain injury (mVBI) is a common pathological correlate of vascular contributions to cognitive impairment and dementia (VCID) that leads to white matter (WM) injury (WMI). VCID appears to arise from chronic recurrent white matter ischemia that triggers oxidative stress and an increase in total oligodendrocyte lineage cells. We hypothesized that mVBI involves vasodilator dysfunction of white matter penetrating arterioles and aberrant oligodendrocyte progenitor cell (OPC) responses to WMI. METHODS We analyzed cases of mVBI with low Alzheimer's disease neuropathological change in prefrontal cortex WM from rapid autopsies in a population-based cohort where VCID frequently occurs. Arteriolar vasodilator function was quantified by videomicroscopy. OPC maturation was quantified using lineage specific markers. RESULTS Acetylcholine-mediated arteriolar dilation in mVBI was significantly reduced in WM penetrators relative to pial arterioles. Astrogliosis-defined WMI was positively associated with increased OPCs and was negatively associated with decreased mature oligodendrocytes. INTERPRETATION Selectively impaired vasodilator function of WM penetrating arterioles in mVBI occurs in association with aberrant differentiation of OPCs in WMI, which supports that myelination disturbances in VCID are related to disrupted maturation of myelinating oligodendrocytes. Ann Neurol 2018;83:142-152.
Collapse
Affiliation(s)
- Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Dieter D. Brandner
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon 97239
| | - Phuong Le
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon 97239
| | - David W. McNeal
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon 97239
| | - Xi Gong
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon 97239
| | - Huijuan Dou
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - David J Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA 30912
| | - Allison Beller
- Department of Pathology, University of Washington, Seattle, Washington
| | - Thuan Ngyuen
- Department of Preventive Medicine, Oregon Health & Science University, Portland, Oregon 97239
| | | | - Thomas J. Montine
- Department of Pathology, University of Washington, Seattle, Washington
| | - C. Dirk Keene
- Department of Pathology, University of Washington, Seattle, Washington
| | - Stephen A. Back
- Department of Pediatrics, Oregon Health & Science University, Portland, Oregon 97239
- Department of Neurology, Oregon Health & Science University, Portland, Oregon 97239
| |
Collapse
|
34
|
Bhatta A, Yao L, Xu Z, Toque HA, Chen J, Atawia RT, Fouda AY, Bagi Z, Lucas R, Caldwell RB, Caldwell RW. Obesity-induced vascular dysfunction and arterial stiffening requires endothelial cell arginase 1. Cardiovasc Res 2017; 113:1664-1676. [PMID: 29048462 PMCID: PMC6410953 DOI: 10.1093/cvr/cvx164] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 03/16/2017] [Accepted: 08/09/2017] [Indexed: 02/04/2023] Open
Abstract
AIMS Elevation of arginase activity has been linked to vascular dysfunction in diabetes and hypertension by a mechanism involving decreased nitric oxide (NO) bioavailability due to L-arginine depletion. Excessive arginase activity also can drive L-arginine metabolism towards the production of ornithine, polyamines, and proline, promoting proliferation of vascular smooth muscle cells and collagen formation, leading to perivascular fibrosis. We hypothesized that there is a specific involvement of arginase 1 expression within the vascular endothelial cells in this pathology. METHODS AND RESULTS To test this proposition, we used models of type 2 diabetes and metabolic syndrome. Studies were performed using wild type (WT), endothelial-specific arginase 1 knockout (EC-A1-/-) and littermate controls(A1con) mice fed high fat-high sucrose (HFHS) or normal diet (ND) for 6 months and isolated vessels exposed to palmitate-high glucose (PA/HG) media. Some WT mice or isolated vessels were treated with an arginase inhibitor, ABH [2-(S)-amino-6-boronohexanoic acid. In WT mice, the HFHS diet promoted increases in body weight, fasting blood glucose, and post-prandial insulin levels along with arterial stiffening and fibrosis, elevated blood pressure, decreased plasma levels of L-arginine, and elevated L-ornithine. The HFHS diet or PA/HG treatment also induced increases in vascular arginase activity along with oxidative stress, reduced vascular NO levels, and impaired endothelial-dependent vasorelaxation. All of these effects except obesity and hypercholesterolemia were prevented or significantly reduced by endothelial-specific deletion of arginase 1 or ABH treatment. CONCLUSION Vascular dysfunctions in diet-induced obesity are prevented by deletion of arginase 1 in vascular endothelial cells or arginase inhibition. These findings indicate that upregulation of arginase 1 expression/activity in vascular endothelial cells has an integral role in diet-induced cardiovascular dysfunction and metabolic syndrome.
Collapse
MESH Headings
- Animals
- Arginase/antagonists & inhibitors
- Arginase/genetics
- Arginase/metabolism
- Arginine/blood
- Blood Glucose/metabolism
- Blood Pressure
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/physiopathology
- Diabetes Mellitus, Experimental/prevention & control
- Diabetes Mellitus, Type 2/enzymology
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/physiopathology
- Diabetes Mellitus, Type 2/prevention & control
- Diet, High-Fat
- Dietary Sucrose
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/enzymology
- Endothelium, Vascular/pathology
- Endothelium, Vascular/physiopathology
- Enzyme Inhibitors/pharmacology
- Fibrosis
- Genetic Predisposition to Disease
- Insulin/blood
- Male
- Metabolic Syndrome/enzymology
- Metabolic Syndrome/genetics
- Metabolic Syndrome/physiopathology
- Metabolic Syndrome/prevention & control
- Mice, Inbred C57BL
- Mice, Knockout
- Nitric Oxide/metabolism
- Obesity/drug therapy
- Obesity/enzymology
- Obesity/genetics
- Obesity/physiopathology
- Ornithine/blood
- Oxidative Stress
- Phenotype
- Signal Transduction
- Vascular Diseases/enzymology
- Vascular Diseases/genetics
- Vascular Diseases/physiopathology
- Vascular Diseases/prevention & control
- Vascular Stiffness/drug effects
- Vasodilation
Collapse
Affiliation(s)
- Anil Bhatta
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Lin Yao
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- School of Pharmaceutical Sciences, South China Research Centre for
Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou 510006, PR
China
| | - Zhimin Xu
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Reem T. Atawia
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
| | - Abdelrahman Y. Fouda
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Zsolt Bagi
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
- Department of Medicine, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| | - Ruth B. Caldwell
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta
University, Augusta, GA 30912, USA
- Veterans Administration Medical Centre, Augusta, GA 30912, USA
| | - Robert W. Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia,
Augusta University, Augusta, GA 30912, USA
- Vascular Biology Centre, Medical College of Georgia, Augusta University,
Augusta, GA 30912, USA
| |
Collapse
|
35
|
Yao L, Bhatta A, Xu Z, Chen J, Toque HA, Chen Y, Xu Y, Bagi Z, Lucas R, Huo Y, Caldwell RB, Caldwell RW. Obesity-induced vascular inflammation involves elevated arginase activity. Am J Physiol Regul Integr Comp Physiol 2017; 313:R560-R571. [PMID: 28835451 PMCID: PMC5792147 DOI: 10.1152/ajpregu.00529.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 08/08/2017] [Accepted: 08/08/2017] [Indexed: 01/29/2023]
Abstract
Obesity-induced vascular dysfunction involves pathological remodeling of the visceral adipose tissue (VAT) and increased inflammation. Our previous studies showed that arginase 1 (A1) in endothelial cells (ECs) is critically involved in obesity-induced vascular dysfunction. We tested the hypothesis that EC-A1 activity also drives obesity-related VAT remodeling and inflammation. Our studies utilized wild-type and EC-A1 knockout (KO) mice made obese by high-fat/high-sucrose (HFHS) diet. HFHS diet induced increases in body weight, fasting blood glucose, and VAT expansion. This was accompanied by increased arginase activity and A1 expression in vascular ECs and increased expression of tumor necrosis factor-α (TNF-α), monocyte chemoattractant protein-1 (MCP-1), interleukin-10 (IL-10), vascular cell adhesion molecule-1 (VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) mRNA and protein in both VAT and ECs. HFHS also markedly increased circulating inflammatory monocytes and VAT infiltration by inflammatory macrophages, while reducing reparative macrophages. Additionally, adipocyte size and fibrosis increased and capillary density decreased in VAT. These effects of HFHS, except for weight gain and hyperglycemia, were prevented or reduced in mice lacking EC-A1 or treated with the arginase inhibitor 2-(S)-amino-6-boronohexanoic acid (ABH). In mouse aortic ECs, exposure to high glucose (25 mM) and Na palmitate (200 μM) reduced nitric oxide production and increased A1, TNF-α, VCAM-1, ICAM-1, and MCP-1 mRNA, and monocyte adhesion. Knockout of EC-A1 or ABH prevented these effects. HFHS diet-induced VAT inflammation is mediated by EC-A1 expression/activity. Limiting arginase activity is a possible therapeutic means of controlling obesity-induced vascular and VAT inflammation.
Collapse
Affiliation(s)
- Lin Yao
- School of Pharmaceutical Sciences, South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Anil Bhatta
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zhimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jijun Chen
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yongjun Chen
- South China Research Center for Acupuncture and Moxibustion, Guangzhou University of Chinese Medicine, Guangzhou, People's Republic of China
| | - Yimin Xu
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zsolt Bagi
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Rudolf Lucas
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Medicine, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Yuqing Huo
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Ruth B Caldwell
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
- Department of Cell Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
- Veterans Administration Medical Center, Augusta, Georgia; and
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia;
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
36
|
Abstract
The nitric oxide-cyclic guanosine monophosphate (NO-cGMP) pathway plays a key role in regulating cardiovascular homeostasis, and genetic variants allocated to NO-cGMP pathway genes, leading to NO-cGMP deficiency, may influence the prevalence or course of cardiovascular disease. NO-cGMP deficiency can be caused by nitric oxide synthase substrate deficiency, substrate competition, defects, or uncoupling; endogenous inhibitors of nitric oxide synthase; decreased cGMP production; or increased cGMP degradation. This review presents evidence supporting the role of NO-cGMP deficiency in cardiovascular disease, including findings from genetic association studies for particular polymorphisms, haplotypes, and racial disparities. NO-cGMP pathway components including arginases, guanosine-5'-triphosphate cyclohydrolase 1, nitric oxide synthase, dimethylarginine dimethylaminohydrolases, soluble guanylyl cyclase, protein kinase G, phosphodiesterase 5, and natriuretic peptides will be discussed.
Collapse
Affiliation(s)
| | - Sven Moosmang
- Bayer AG, Experimental Medicine CV/HEM, Wuppertal, Germany
| | | |
Collapse
|
37
|
Förstermann U, Xia N, Li H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ Res 2017; 120:713-735. [DOI: 10.1161/circresaha.116.309326] [Citation(s) in RCA: 692] [Impact Index Per Article: 98.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 12/19/2016] [Accepted: 12/26/2016] [Indexed: 12/13/2022]
Abstract
Major reactive oxygen species (ROS)–producing systems in vascular wall include NADPH (reduced form of nicotinamide adenine dinucleotide phosphate) oxidase, xanthine oxidase, the mitochondrial electron transport chain, and uncoupled endothelial nitric oxide (NO) synthase. ROS at moderate concentrations have important signaling roles under physiological conditions. Excessive or sustained ROS production, however, when exceeding the available antioxidant defense systems, leads to oxidative stress. Animal studies have provided compelling evidence demonstrating the roles of vascular oxidative stress and NO in atherosclerosis. All established cardiovascular risk factors such as hypercholesterolemia, hypertension, diabetes mellitus, and smoking enhance ROS generation and decrease endothelial NO production. Key molecular events in atherogenesis such as oxidative modification of lipoproteins and phospholipids, endothelial cell activation, and macrophage infiltration/activation are facilitated by vascular oxidative stress and inhibited by endothelial NO. Atherosclerosis develops preferentially in vascular regions with disturbed blood flow (arches, branches, and bifurcations). The fact that these sites are associated with enhanced oxidative stress and reduced endothelial NO production is a further indication for the roles of ROS and NO in atherosclerosis. Therefore, prevention of vascular oxidative stress and improvement of endothelial NO production represent reasonable therapeutic strategies in addition to the treatment of established risk factors (hypercholesterolemia, hypertension, and diabetes mellitus).
Collapse
Affiliation(s)
- Ulrich Förstermann
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Ning Xia
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| | - Huige Li
- From the Department of Pharmacology, Johannes Gutenberg University Medical Center, Mainz, Germany (U.F., N.X., H.L.); Center for Translational Vascular Biology (CTVB), Johannes Gutenberg University Medical Center, Mainz, Germany (H.L.); and German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany (H.L.)
| |
Collapse
|
38
|
Pudlo M, Demougeot C, Girard-Thernier C. Arginase Inhibitors: A Rational Approach Over One Century. Med Res Rev 2016; 37:475-513. [DOI: 10.1002/med.21419] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 09/14/2016] [Accepted: 09/22/2016] [Indexed: 12/28/2022]
Affiliation(s)
- Marc Pudlo
- PEPITE - EA4267; University Bourgogne Franche-Comté; Besançon France
| | - Céline Demougeot
- PEPITE - EA4267; University Bourgogne Franche-Comté; Besançon France
| | | |
Collapse
|
39
|
Santiago E, Martínez MP, Climent B, Muñoz M, Briones AM, Salaices M, García-Sacristán A, Rivera L, Prieto D. Augmented oxidative stress and preserved vasoconstriction induced by hydrogen peroxide in coronary arteries in obesity: role of COX-2. Br J Pharmacol 2016; 173:3176-3195. [PMID: 27535007 DOI: 10.1111/bph.13579] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 08/02/2016] [Accepted: 08/03/2016] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND AND PURPOSE Oxidative stress plays a key role in the vascular and metabolic abnormalities associated with obesity. Herein, we assessed whether obesity can increase coronary vasoconstriction induced by hydrogen peroxide (H2 O2 ) and the signalling pathways involving COX-2 and superoxide (O2.- ) generation. EXPERIMENTAL APPROACH Contractile responses to H2 O2 and O2.- generation were measured in coronary arteries from genetically obese Zucker rats (OZR) and compared to lean Zucker rats (LZR). KEY RESULTS Both basal and H2 O2 -stimulated O2.- production were enhanced in coronary arteries from OZR, but H2 O2 -induced vasoconstriction was unchanged. The selective COX-2 inhibitor NS398 significantly reduced H2 O2 -induced contractions in endothelium-denuded arteries from LZR and OZR, but only in endothelium-intact arteries from LZR. PGI2 (IP) receptor antagonism modestly reduced the vasoconstrictor action of H2 O2 while antagonism of the PGE2 receptor 4 (EP4 ) enhanced H2 O2 contractions in arteries from OZR but not LZR. Basal release of COX-2-derived PGE2 was higher in coronary arteries from OZR where the selective agonist of EP4 receptors TCS 2519 evoked potent relaxations. COX-2 was up-regulated after acute exposure to H2 O2 in coronary endothelium and vascular smooth muscle (VSM) and inhibition of COX-2 markedly reduced H2 O2 -elicited O2.- generation in coronary arteries and myocardium. Expression of Nox subunits in VSM and NADPH-stimulated O2.- generation was enhanced and contributed to H2 O2 vasoconstriction in arteries from obese rats. CONCLUSION AND IMPLICATIONS COX-2 contributes to cardiac oxidative stress and to the endothelium-independent O2.- -mediated coronary vasoconstriction induced by H2 O2 in obesity, which is offset by the release of COX-2-derived endothelial PGE2 acting on EP4 vasodilator receptors.
Collapse
Affiliation(s)
- Elvira Santiago
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Maria Pilar Martínez
- Departamento de Anatomía and Anatomía Patológica Comparadas, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Belén Climent
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Mercedes Muñoz
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Ana María Briones
- Departamento de Farmacología, Facultad de Medicina, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Mercedes Salaices
- Departamento de Farmacología, Facultad de Medicina, Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Universidad Autónoma de Madrid, Madrid, Spain
| | - Albino García-Sacristán
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis Rivera
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Dolores Prieto
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain.
| |
Collapse
|
40
|
Forte M, Conti V, Damato A, Ambrosio M, Puca AA, Sciarretta S, Frati G, Vecchione C, Carrizzo A. Targeting Nitric Oxide with Natural Derived Compounds as a Therapeutic Strategy in Vascular Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7364138. [PMID: 27651855 PMCID: PMC5019908 DOI: 10.1155/2016/7364138] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/29/2016] [Revised: 07/30/2016] [Accepted: 08/01/2016] [Indexed: 12/20/2022]
Abstract
Within the family of endogenous gasotransmitters, nitric oxide (NO) is the smallest gaseous intercellular messenger involved in the modulation of several processes, such as blood flow and platelet aggregation control, essential to maintain vascular homeostasis. NO is produced by nitric oxide synthases (NOS) and its effects are mediated by cGMP-dependent or cGMP-independent mechanisms. Growing evidence suggests a crosstalk between the NO signaling and the occurrence of oxidative stress in the onset and progression of vascular diseases, such as hypertension, heart failure, ischemia, and stroke. For these reasons, NO is considered as an emerging molecular target for developing therapeutic strategies for cardio- and cerebrovascular pathologies. Several natural derived compounds, such as polyphenols, are now proposed as modulators of NO-mediated pathways. The aim of this review is to highlight the experimental evidence on the involvement of nitric oxide in vascular homeostasis focusing on the therapeutic potential of targeting NO with some natural compounds in patients with vascular diseases.
Collapse
Affiliation(s)
- Maurizio Forte
- IRCCS Neuromed, Vascular Physiopathology Unit, Pozzilli, Italy
| | - Valeria Conti
- Università degli Studi di Salerno, Medicine, Surgery and Dentistry, Baronissi, Italy
| | - Antonio Damato
- IRCCS Neuromed, Vascular Physiopathology Unit, Pozzilli, Italy
| | | | - Annibale A. Puca
- Università degli Studi di Salerno, Medicine, Surgery and Dentistry, Baronissi, Italy
- IRCCS Multimedica, Milan, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Vascular Physiopathology Unit, Pozzilli, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Giacomo Frati
- IRCCS Neuromed, Vascular Physiopathology Unit, Pozzilli, Italy
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy
| | - Carmine Vecchione
- IRCCS Neuromed, Vascular Physiopathology Unit, Pozzilli, Italy
- Università degli Studi di Salerno, Medicine, Surgery and Dentistry, Baronissi, Italy
| | - Albino Carrizzo
- IRCCS Neuromed, Vascular Physiopathology Unit, Pozzilli, Italy
| |
Collapse
|
41
|
Ciuntu A. Role of Metabolites of Nitric Oxide and Arginase in the Pathogenesis of Glomerulonephritis. CURRENT HEALTH SCIENCES JOURNAL 2016; 42:221-225. [PMID: 30581575 PMCID: PMC6269603 DOI: 10.12865/chsj.42.03.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/01/2016] [Indexed: 11/18/2022]
Abstract
PURPOSE The aim of the study is to assess the level of nitric oxide metabolites and arginase in the urine of children with glomerulonephritis depending on clinical evolutional stages of the disease. MATERIALS AND METHODS The prospective study included 65 children with primary glomerulonephritis, 25 children with steroid-sensitive nephrotic syndrome (SSNS) and 20 children with steroid-resistant nephrotic syndrome (SRNS), 20 children with mixed form of chronic glomerulonephritis(CGN). RESULTS Thus in the SRNS group, during relapse period the concentration of NO metabolites in urine was increased by 4,2 times, while in SSNS by 3,0 times in comparison with the control group. The concentration of NO metabolites in the urine increased by 4,8 times during relapse CGN mixed form in comparison to the control values. During remission, the levels of NO metabolites in the urine remain increased in both groups. In relapse of SSNS arginase levels in the urine increased by 4,5 times in comparation to SRNS, thus the concentration of arginase was reduced. During remission period arginase levels in the urine were practically reduced to the levels of the control group. In the mixed form of CGN, relapse period arginase levels in the urine were increased by 2,9 times and during remission were decreased by almost 1,9 times in comparision to the control group. CONCLUSIONS Assesment of NO metabolites and arginase in urine can be used as a diagnostic method in order to monitor renal disease process, evolution and effectiveness of the applied treatment.
Collapse
Affiliation(s)
- A Ciuntu
- Pediatric Department, State University of Medicine and Pharmacy "Nicolae Testemitanu", Chișinău Paediatrics National Institute of Health Care for Mother and Child, Nephrology Unit, R.Moldova
| |
Collapse
|
42
|
Siragusa M, Fleming I. The eNOS signalosome and its link to endothelial dysfunction. Pflugers Arch 2016; 468:1125-1137. [DOI: 10.1007/s00424-016-1839-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 05/10/2016] [Indexed: 12/17/2022]
|
43
|
Adipokine Imbalance in the Pericardial Cavity of Cardiac and Vascular Disease Patients. PLoS One 2016; 11:e0154693. [PMID: 27139713 PMCID: PMC4854456 DOI: 10.1371/journal.pone.0154693] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/18/2016] [Indexed: 01/09/2023] Open
Abstract
Aim Obesity and especially hypertrophy of epicardial adipose tissue accelerate coronary atherogenesis. We aimed at comparing levels of inflammatory and atherogenic hormones from adipose tissue in the pericardial fluid and circulation of cardiovascular disease patients. Methods and Results Venous plasma (P) and pericardial fluid (PF) were obtained from elective cardiothoracic surgery patients (n = 37). Concentrations of leptin, adipocyte fatty acid-binding protein (A-FABP) and adiponectin (APN) were determined by enzyme-linked immunosorbent assays (ELISA). The median concentration of leptin in PF (4.3 (interquartile range: 2.8–9.1) μg/L) was comparable to that in P (5.9 (2.2–11) μg/L) and these were significantly correlated to most of the same patient characteristics. The concentration of A-FABP was markedly higher (73 (28–124) versus 8.4 (5.2–14) μg/L) and that of APN was markedly lower (2.8 (1.7–4.2) versus 13 (7.2–19) mg/L) in PF compared to P. APN in PF was unlike in P not significantly related to age, body mass index, plasma triglycerides or coronary artery disease. PF levels of APN, but not A-FABP, were related to the size of paracardial adipocytes. PF levels of APN and A-FABP were not related to the immunoreactivity of paracardial adipocytes for these proteins. Conclusion In cardiac and vascular disease patients, PF is enriched in A-FABP and poor in APN. This adipokine microenvironment is more likely determined by the heart than by the circulation or paracardial adipose tissue.
Collapse
|
44
|
Xiong S, Wang P, Ma L, Gao P, Gong L, Li L, Li Q, Sun F, Zhou X, He H, Chen J, Yan Z, Liu D, Zhu Z. Ameliorating Endothelial Mitochondrial Dysfunction Restores Coronary Function via Transient Receptor Potential Vanilloid 1-Mediated Protein Kinase A/Uncoupling Protein 2 Pathway. Hypertension 2015; 67:451-60. [PMID: 26667415 DOI: 10.1161/hypertensionaha.115.06223] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 11/19/2015] [Indexed: 01/02/2023]
Abstract
Coronary heart disease arising from atherosclerosis is a leading cause of cardiogenic death worldwide. Mitochondria are the principal source of reactive oxygen species (ROS), and defective oxidative phosphorylation by the mitochondrial respiratory chain contributes to ROS generation. Uncoupling protein 2 (UCP2), an adaptive antioxidant defense factor, protects against mitochondrial ROS-induced endothelial dysfunction in atherosclerosis. The activation of transient receptor potential vanilloid 1 (TRPV1) attenuates vascular dysfunction. Therefore, whether TRPV1 activation antagonizes coronary lesions by alleviating endothelial mitochondrial dysfunction and enhancing the activity of the protein kinase A/UCP2 pathway warrants examination. ApoE(-/-), ApoE(-/-)/TRPV1(-/-), and ApoE(-/-)/UCP2(-/-) mice were fed standard chow, a high-fat diet (HFD), or the HFD plus 0.01% capsaicin. HFD intake profoundly impaired coronary vasodilatation and myocardial perfusion and shortened the survival duration of ApoE(-/-) mice. TRPV1 or UCP2 deficiency exacerbated HFD-induced coronary dysfunction and was associated with increased ROS generation and reduced nitric oxide production in the endothelium. The activation of TRPV1 by capsaicin upregulated UCP2 expression via protein kinase A phosphorylation, thereby alleviating endothelial mitochondrial dysfunction and inhibiting mitochondrial ROS generation. In vivo, dietary capsaicin supplementation enhanced coronary relaxation and prolonged the survival duration of HFD-fed ApoE(-/-) mice. These effects were not observed in ApoE(-/-) mice lacking the TRPV1 or UCP2 gene. The upregulation of protein kinase A /UCP2 via TRPV1 activation ameliorates coronary dysfunction and prolongs the lifespan of atherosclerotic mice by ameliorating endothelial mitochondrial dysfunction. Dietary capsaicin supplementation may represent a promising intervention for the primary prevention of coronary heart disease.
Collapse
Affiliation(s)
- Shiqiang Xiong
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Peijian Wang
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Liqun Ma
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Peng Gao
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Liuping Gong
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Li Li
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Qiang Li
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Fang Sun
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Xunmei Zhou
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Hongbo He
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Jing Chen
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Zhencheng Yan
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China
| | - Daoyan Liu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China.
| | - Zhiming Zhu
- From the Department of Hypertension and Endocrinology, Center for Hypertension and Metabolic Diseases, Daping Hospital, Third Military Medical University, Chongqing Institute of Hypertension, Chongqing, China.
| |
Collapse
|
45
|
Pernow J, Kiss A, Tratsiakovich Y, Climent B. Tissue-specific up-regulation of arginase I and II induced by p38 MAPK mediates endothelial dysfunction in type 1 diabetes mellitus. Br J Pharmacol 2015; 172:4684-98. [PMID: 26140333 PMCID: PMC4594272 DOI: 10.1111/bph.13242] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 05/13/2015] [Accepted: 06/26/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Emerging evidence suggests a selective up-regulation of arginase I in diabetes causing coronary artery disease; however, the mechanisms behind this up-regulation are still unknown. Activated p38 MAPK has been reported to increase arginase II in various cardiovascular diseases. We therefore tested the role of p38 MAPK in the regulation of arginase I and II expression and its effect on endothelial dysfunction in diabetes mellitus. EXPERIMENTAL APPROACH Endothelial function was determined in septal coronary (SCA), left anterior descending coronary (LAD) and mesenteric (MA) arteries from healthy and streptozotocin-induced diabetic Wistar rats by wire myographs. Arginase activity and protein levels of arginase I, II, phospho-p38 MAPK and phospho-endothelial NOS (eNOS) (Ser(1177) ) were determined in these arteries from diabetic and healthy rats treated with a p38 MAPK inhibitor in vivo. KEY RESULTS Diabetic SCA and MA displayed impaired endothelium-dependent relaxation, which was prevented by arginase and p38 MAPK inhibition while LAD relaxation was not affected. Arginase I, phospho-p38 MAPK and eNOS protein expression was increased in diabetic coronary arteries. In diabetic MA, however, increased expression of arginase II and phospho-p38 MAPK, increased arginase activity and decreased expression of eNOS were observed. All these effects were reversed by p38 MAPK inhibition. CONCLUSIONS AND IMPLICATIONS Diabetes-induced activation of p38 MAPK causes endothelial dysfunction via selective up-regulation of arginase I expression in coronary arteries and arginase II expression in MA. Therefore, regional differences appear to exist in the arginase isoforms contributing to endothelial dysfunction in type 1 diabetes mellitus.
Collapse
Affiliation(s)
- J Pernow
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - A Kiss
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Y Tratsiakovich
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - B Climent
- Division of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense, Madrid, Spain
| |
Collapse
|
46
|
Abstract
OBJECTIVE Several environmental factors including hypoxia have been reported to contribute to oxidative stress in individuals living in the highlands. However, little is known about the role of oxidized low-density lipoprotein (ox-LDL) among community-dwelling elderly in the Qinghai-Tibet plateau. METHODS The study population comprised 168 community-dwelling elderly subjects aged 60 years or older (male to female ratio, 70:98; mean age, 65.8 years) living in Haiyan County, located 3000 to 3200 m above sea level, 30 km northwest of Xining, Qinghai. The subjects were volunteers who joined a Comprehensive Geriatric Assessment. Plasma ox-LDL was measured in 168 community-dwelling elderly subjects aged 60 years or older (23 Tibetans and 145 Hans) with a monoclonal antibody-based enzyme-linked immunosorbent assay. RESULTS Mean ox-LDL level was higher among Tibetan elderly than Han elderly (Tibetan, 79.0 ± 29.6 U/L; Han, 62.8 ± 23.5 U/L; P = .003). Tibetan ethnicity was significantly associated with ox-LDL levels after adjusting for LDL cholesterol levels. In addition, high ox-LDL levels (≥70 U/L) were significantly associated with a homeostasis model assessment insulin resistance index of at least 1.6 (odds ratio [OR], 2.82; 95% confidence interval [95% CI], 1.11 to 7.15; P = .029) and ankle brachial pressure index of less than 1.0 (OR, 4.85; 95% CI, 1.14 to 10.00; P = .028), after adjusting for age, sex, and ethnicity. CONCLUSIONS Our findings support the hypothesis that ox-LDL levels are higher among Tibetan elderly highlanders compared with those among Han elderly. As ox-LDL levels can affect insulin resistance and arteriosclerosis, further research is needed to determine how oxidative stress influences the health situation among elderly individuals at high altitudes.
Collapse
|
47
|
Caldwell RB, Toque HA, Narayanan SP, Caldwell RW. Arginase: an old enzyme with new tricks. Trends Pharmacol Sci 2015; 36:395-405. [PMID: 25930708 PMCID: PMC4461463 DOI: 10.1016/j.tips.2015.03.006] [Citation(s) in RCA: 207] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 03/23/2015] [Accepted: 03/30/2015] [Indexed: 01/05/2023]
Abstract
Arginase has roots in early life-forms. It converts L-arginine to urea and ornithine. The former provides protection against NH3; the latter serves to stimulate cell growth and other physiological functions. Excessive arginase activity in mammals has been associated with cardiovascular and nervous system dysfunction and disease. Two relevant aspects of this elevated activity may be involved in these disease states. First, excessive arginase activity reduces the supply of L-arginine needed by nitric oxide (NO) synthase to produce NO. Second, excessive production of ornithine leads to vascular structural problems and neural toxicity. Recent research has identified inflammatory agents and reactive oxygen species (ROS) as drivers of this pathologic elevation of arginase activity and expression. We review the involvement of arginase in cardiovascular and nervous system dysfunction, and discuss potential therapeutic interventions targeting excess arginase.
Collapse
Affiliation(s)
- Ruth B. Caldwell
- VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA
- Vision Discovery Institute, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - Haroldo A. Toque
- Department of Pharmacology & Toxicology, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - S. Priya Narayanan
- Vision Discovery Institute, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Vascular Biology Center, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Department of Occupational Therapy, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| | - R. William Caldwell
- Vision Discovery Institute, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
- Department of Pharmacology & Toxicology, School of Allied Health Sciences, Medical College of Georgia, Georgia Regents University, 1459 Laney Walker Boulevard, Augusta, 30912, USA
| |
Collapse
|
48
|
Henno P, Maurey C, Le Pimpec-Barthes F, Devillier P, Delclaux C, Israël-Biet D. Is arginase a potential drug target in tobacco-induced pulmonary endothelial dysfunction? Respir Res 2015; 16:46. [PMID: 25889611 PMCID: PMC4391310 DOI: 10.1186/s12931-015-0196-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 02/25/2015] [Indexed: 11/29/2022] Open
Abstract
Background Tobacco-induced pulmonary vascular disease is partly driven by endothelial dysfunction. The bioavailability of the potent vasodilator nitric oxide (NO) depends on competition between NO synthase-3 (NOS3) and arginases for their common substrate (L-arginine). We tested the hypothesis whereby tobacco smoking impairs pulmonary endothelial function via upregulation of the arginase pathway. Methods Endothelium-dependent vasodilation in response to acetylcholine (Ach) was compared ex vivo for pulmonary vascular rings from 29 smokers and 10 never-smokers. The results were expressed as a percentage of the contraction with phenylephrine. We tested the effects of L-arginine supplementation, arginase inhibition (by N(omega)-hydroxy-nor-l-arginine, NorNOHA) and NOS3 induction (by genistein) on vasodilation. Protein levels of NOS3 and arginases I and II in the pulmonary arteries were quantified by Western blotting. Results Overall, vasodilation was impaired in smokers (relative to controls; p < 0.01). Eleven of the 29 smokers (the ED+ subgroup) displayed endothelial dysfunction (defined as the absence of a relaxant response to Ach), whereas 18 (the ED− subgroup) had normal vasodilation. The mean responses to 10−4 M Ach were −23 ± 10% and 31 ± 4% in the ED+ and ED− subgroups, respectively (p < 0.01). Supplementation with L- arginine improved endothelial function in the ED+ subgroup (−4 ± 10% vs. -32 ± 10% in the presence and absence of L- arginine, respectively; p = 0.006), as did arginase inhibition (18 ± 9% vs. -1 ± 9%, respectively; p = 0.0002). Arginase I protein was overexpressed in ED+ samples, whereas ED+ and ED− samples did not differ significantly in terms of NOS3 expression. Treatment with genistein did not significantly improve endothelial function in ED+ samples. Conclusion Overexpression and elevated activity of arginase I are involved in tobacco-induced pulmonary endothelial dysfunction.
Collapse
Affiliation(s)
- Priscilla Henno
- Sorbonne Universités, UPMC Université Paris 06, Paris, France. .,Département Physiologie-Algologie-Somnologie, Unité Fonctionnelle de Somnologie et Fonction Respiratoire, AP-HP, Hôpital Saint Antoine, 75012, Paris, France. .,Laboratoire de Pharmacologie Respiratoire UPRES EA 220, Hôpital Foch, 92150, Suresnes, France.
| | - Christelle Maurey
- Ecole Nationale Vétérinaire d'Alfort, Unité de Médecine, Université Paris-Est, 94700, Maisons-Alfort, France.
| | - Françoise Le Pimpec-Barthes
- Sorbonne Paris Cité, Université Paris-Descartes, Paris, France. .,Service de Chirurgie Thoracique, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France.
| | - Philippe Devillier
- Laboratoire de Pharmacologie Respiratoire UPRES EA 220, Hôpital Foch, 92150, Suresnes, France. .,Université Versailles Saint-Quentin en Yvelines, UFR Sciences de la Santé Simone Veil, Montigny le Bretonneux, France.
| | - Christophe Delclaux
- Sorbonne Paris Cité, Université Paris-Descartes, Paris, France. .,Service de Physiologie, Explorations Fonctionnelles Respiratoires et du Sommeil, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France.
| | - Dominique Israël-Biet
- Sorbonne Paris Cité, Université Paris-Descartes, Paris, France. .,Service de Pneumologie, AP-HP, Hôpital Européen Georges Pompidou, 75015, Paris, France.
| |
Collapse
|
49
|
Johnson JM, Minson CT, Kellogg DL. Cutaneous vasodilator and vasoconstrictor mechanisms in temperature regulation. Compr Physiol 2014; 4:33-89. [PMID: 24692134 DOI: 10.1002/cphy.c130015] [Citation(s) in RCA: 241] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In this review, we focus on significant developments in our understanding of the mechanisms that control the cutaneous vasculature in humans, with emphasis on the literature of the last half-century. To provide a background for subsequent sections, we review methods of measurement and techniques of importance in elucidating control mechanisms for studying skin blood flow. In addition, the anatomy of the skin relevant to its thermoregulatory function is outlined. The mechanisms by which sympathetic nerves mediate cutaneous active vasodilation during whole body heating and cutaneous vasoconstriction during whole body cooling are reviewed, including discussions of mechanisms involving cotransmission, NO, and other effectors. Current concepts for the mechanisms that effect local cutaneous vascular responses to local skin warming and cooling are examined, including the roles of temperature sensitive afferent neurons as well as NO and other mediators. Factors that can modulate control mechanisms of the cutaneous vasculature, such as gender, aging, and clinical conditions, are discussed, as are nonthermoregulatory reflex modifiers of thermoregulatory cutaneous vascular responses.
Collapse
Affiliation(s)
- John M Johnson
- Department of Physiology, University of Texas Health Science Center at San Antonio, San Antonio, Texas
| | | | | |
Collapse
|
50
|
You H, Gao T, Cooper TK, Morris SM, Awad AS. Diabetic nephropathy is resistant to oral L-arginine or L-citrulline supplementation. Am J Physiol Renal Physiol 2014; 307:F1292-301. [PMID: 25320354 DOI: 10.1152/ajprenal.00176.2014] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Our recent publication showed that pharmacological blockade of arginases confers kidney protection in diabetic nephropathy via a nitric oxide (NO) synthase (NOS)3-dependent mechanism. Arginase competes with endothelial NOS (eNOS) for the common substrate L-arginine. Lack of L-arginine results in reduced NO production and eNOS uncoupling, which lead to endothelial dysfunction. Therefore, we hypothesized that L-arginine or L-citrulline supplementation would ameliorate diabetic nephropathy. DBA mice injected with multiple low doses of vehicle or streptozotocin (50 mg/kg ip for 5 days) were provided drinking water with or without L-arginine (1.5%, 6.05 g·kg(-1)·day(-1)) or L-citrulline (1.66%, 5.73 g·kg(-1)·day(-1)) for 9 wk. Nonsupplemented diabetic mice showed significant increases in albuminuria, blood urea nitrogen, glomerular histopathological changes, kidney macrophage recruitment, kidney TNF-α and fibronectin mRNA expression, kidney arginase activity, kidney arginase-2 protein expression, and urinary oxidative stress along with a significant reduction of nephrin and eNOS protein expression and kidney nitrite + nitrate compared with normal mice after 9 wk of diabetes. Surprisingly, L-arginine or L-citrulline supplementation in diabetic mice did not affect any of these parameters despite greatly increasing kidney and plasma arginine levels. These findings demonstrate that chronic L-arginine or L-citrulline supplementation does not prevent or reduce renal injury in a model of type 1 diabetes.
Collapse
Affiliation(s)
- Hanning You
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Ting Gao
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania
| | - Timothy K Cooper
- Department of Comparative Medicine, Penn State University College of Medicine, Hershey, Pennsylvania; and
| | - Sidney M Morris
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Alaa S Awad
- Division of Nephrology, Department of Medicine, Penn State University College of Medicine, Hershey, Pennsylvania;
| |
Collapse
|