1
|
Thakur MR, Tupe RS. l-Arginine: A multifaceted regulator of diabetic cardiomyopathy. Biochem Biophys Res Commun 2025; 761:151720. [PMID: 40186920 DOI: 10.1016/j.bbrc.2025.151720] [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: 02/14/2025] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
In diabetes mellitus, dysregulated glucose and lipid metabolism lead to diabetic cardiomyopathy (DCM) by imparting pathological myocardial remodeling and cellular injury. Accelerated glycation, oxidative stress, and activated inflammatory pathways culminate in cardiac fibrosis and hypertrophy in DCM. The regulatory effects of l-Arginine (L-Arg) have been elucidated in the pathological changes of DCM, including myocardial fibrosis, hypertrophy, and apoptosis, by inhibiting glycation and oxidative stress-induced inflammation. Disturbed L-Arg metabolism and decreased intracellular L-Arg pool are correlated with the progression of DCM; therefore, L-Arg supplementation has been prescribed for various cardiovascular dysfunctions. This review expands the therapeutic potential of L-Arg supplementation in DCM by elucidating its molecular mechanism of action and exploring potential clinical outcomes.
Collapse
Affiliation(s)
- Muskan R Thakur
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India
| | - Rashmi S Tupe
- Symbiosis School of Biological Sciences (SSBS), Symbiosis International (Deemed University) (SIU), Lavale, Pune, 412115, Maharashtra, India.
| |
Collapse
|
2
|
Chen XH, Li L, Ma SY, Ling CQ, Lai JQ, Yao HJ, Guo WB, Yang Y, Zhang Y, Lin WJ, He FZ. Arginase 1 genetic variation is associated with the risk of vascular complications in type 2 diabetes. Biomark Med 2025; 19:63-72. [PMID: 39885775 PMCID: PMC11792801 DOI: 10.1080/17520363.2025.2455924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
OBJECTIVE This study aims to explore the association between arginase 1 (ARG1) genetic variation and susceptibility to type 2 diabetes (T2DM) vascular complications, a primary cause of morbidity and mortality in diabetics. METHODS ARG1, a risk gene for cardiovascular disease, was identified from GEO datasets GSE22255 and GSE58294. The ENCODE database identified four candidate single-nucleotide polymorphism (SNP) loci. Nine hundred ninety-two T2DM patients underwent SNP genotyping, and relevant biochemical markers were tested. Logistic regression analysis calculated the odds ratio (OR) and 95% confidence interval (CI) between ARG1 SNP and diabetic vascular complications. RESULTS Out of 985 patients, 250 had CHD, with the TTTG group accounting for 64/250. This group showed a significant reduction in CHD risk (non-TTTG factor-adjusted OR = 1.61, 95% CI: 1.14-2.29, p = 0.008). The combination of the TTTG group, age, central obesity, and hypertension better predicted CHD risk (Area under the curve = 0.72, p < 0.001). CONCLUSIONS ARG1 polymorphisms significantly impact vascular complications in T2DM patients, implying that ARG1 genetic variation may be a potential prevention and treatment target. TRIAL REGISTRATION ChiCTR1800015661.
Collapse
Affiliation(s)
- X H Chen
- College of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - L Li
- Department of Pharmacy, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - S Y Ma
- Department of Pharmacy, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - C Q Ling
- Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - J Q Lai
- College of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - H J Yao
- College of Pharmacy, Jinan University, Guangzhou, China
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - W B Guo
- The 3rd People's Hospital of Yuxi City, Yuxi, China
| | - Y Yang
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - Y Zhang
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - W J Lin
- Department of Pharmacy, Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| | - F Z He
- Zhuhai People's Hospital, Zhuhai Hospital Affiliated with Jinan University, Zhuhai, China
| |
Collapse
|
3
|
Heuser SK, Li J, Pudewell S, LoBue A, Li Z, Cortese-Krott MM. Biochemistry, pharmacology, and in vivo function of arginases. Pharmacol Rev 2025; 77:100015. [PMID: 39952693 DOI: 10.1124/pharmrev.124.001271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 01/22/2025] Open
Abstract
The enzyme arginase catalyzes the hydrolysis of l-arginine into l-ornithine and urea. The 2 existing isoforms Arg1 and Arg2 exhibit different cellular localizations and metabolic functions. Arginase activity is crucial for nitrogen detoxification in the urea cycle, synthesis of polyamines, and control of l-arginine bioavailability and nitric oxide (NO) production. Despite significant progress in the understanding of the biochemistry and function of arginases, several open questions remain. Recent studies have revealed that the regulation and function of Arg1 and Arg2 are cell type-specific, species-specific, and profoundly different in mice and humans. The main differences are in the distribution and function of Arg1 and Arg2 in immune and erythroid cells. Contrary to what was previously thought, Arg1 activity appears to be only partially related to vascular NO signaling under homeostatic conditions in the vascular wall, but its expression is increased under disease conditions and may be targeted by treatment with arginase inhibitors. Arg2 appears to be mainly a catabolic enzyme involved in the synthesis of l-ornithine, polyamine, and l-proline but may play a putative role in blood pressure control, at least in mice. The immunosuppressive role of arginase-mediated arginine depletion is a promising target for cancer treatment. This review critically revises and discusses the biochemistry, pharmacology, and in vivo function of arginases, focusing on the insights gained from the analysis of cell-specific Arg1 and Arg2 knockout mice and human studies using arginase inhibitors or pegylated recombinant arginase. SIGNIFICANCE STATEMENT: Further basic and translational research is needed to deepen our understanding of the regulation of Arg1 and Arg2 in different cell types in consideration of their localization, species-specificity, and multiple biochemical and physiological roles. This will lead to better pharmacological strategies to target arginase activity in liver, cardiovascular, hematological, immune/infectious diseases, and cancer.
Collapse
Affiliation(s)
- Sophia K Heuser
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Junjie Li
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Silke Pudewell
- Department of Biochemistry and Molecular Biology II, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anthea LoBue
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Zhixin Li
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Cardiovascular Research Institute Düsseldorf, Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
4
|
Amin KN, Rajaguru P, Suzuki T, Sarkar K, Ganesan K, Ramkumar KM. Quantitative proteomic analyses uncover regulatory roles of Nrf2 in human endothelial cells. Cell Stress Chaperones 2023; 28:731-747. [PMID: 37488350 PMCID: PMC10746666 DOI: 10.1007/s12192-023-01366-5] [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: 04/05/2023] [Revised: 06/13/2023] [Accepted: 07/10/2023] [Indexed: 07/26/2023] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2), a transcriptional regulator, is the predominant factor in modulating oxidative stress and other cellular signaling responses. Studies from our lab and others highlighted that activation of the Nrf2 pathway by small molecules improves endothelial function by suppressing oxidative and endoplasmic reticulum (ER) stress. However, the exact mechanisms by which Nrf2 elicits these effects are unknown. In the present study, we developed CRISPR/Cas9-mediated Nrf2 knocked-out human endothelial cells, and proteomic signature was studied using LC-MS/MS. We identified 723 unique proteins, of which 361 proteins were found to be differentially regulated and further screened in the Nrf2ome online database, where we identified a highly interconnected signaling network in which 70 proteins directly interact with Nrf2. These proteins were found to regulate some key cellular and metabolic processes in the regulation actin cytoskeleton, ER stress, angiogenesis, inflammation, Hippo signaling pathway, and epidermal growth factor/fibroblast growth factor (EGF/FGF) signaling pathway. Our findings suggest the role of Nrf2 in maintaining endothelium integrity and its relationship with the crucial cellular processes which help develop novel therapeutics against endothelial dysfunction and its associated complications.
Collapse
Affiliation(s)
- Karan Naresh Amin
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Palanichamy Rajaguru
- Department of Biotechnology, Central University of Tamil Nadu, Tiruvarur, 610005, India
| | - Takayoshi Suzuki
- Division Cellular and Gene Therapy Products, National Institute of Health Sciences, Setagaya-Ku, Tokyo, 158-8501, Japan
| | - Koustav Sarkar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India
| | - Kumar Ganesan
- School of Chinese Medicine, LKS Faculty of Medicine, University of Hong Kong, 10 Sassoon Road, Hong Kong, 999077, China
| | - Kunka Mohanram Ramkumar
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603203, Tamil Nadu, India.
| |
Collapse
|
5
|
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
|
6
|
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: 16] [Impact Index Per Article: 5.3] [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
|
7
|
Buraczynska M, Zakrocka I. Arginase Gene Polymorphism Increases Risk of Diabetic Retinopathy in Type 2 Diabetes Mellitus Patients. J Clin Med 2021; 10:jcm10225407. [PMID: 34830689 PMCID: PMC8620112 DOI: 10.3390/jcm10225407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 12/22/2022] Open
Abstract
Studies have demonstrated that polymorphic variants of arginase 1 gene (ARG1) are involved in human diseases, such as coronary heart disease, hypertension, and diabetes. Our study aimed to investigate the association between ARG1 rs2781666 single nucleotide polymorphism (SNP) and diabetic retinopathy (DR) in type 2 diabetes (T2DM) patients. Polymorphism was genotyped in 740 T2DM patients and 400 healthy individuals. A significant difference in the genotype distribution was observed between the patients and the controls. The T allele and TT genotype were associated with an increased risk of T2DM (OR 1.4, 95% CI 1.14–1.72, p = 0.001 and OR 2.16, 95% CI 1.23–3.80, p = 0.007, respectively). When the T2DM subjects were stratified into DR+ and DR− subgroups, the T allele and TT genotype frequencies were significantly higher in the DR+ group compared to the DR− group, demonstrating OR 1.68 (1.33–2.12), p < 0.0001 and OR 2.39 (1.36–4.18), p = 0.002, respectively. Logistic regression analysis was applied to determine the interaction between the ARG1 genotypes and other risk factors. Only ARG1 rs2781666 SNP was a significant risk predictor of DR (p = 0.003). In conclusion, this is the first report discussing the effect of ARG1 polymorphism on the microvascular complications that are associated with diabetes. Our findings demonstrate that ARG1 rs2781666 SNP is significantly associated with an increased susceptibility to DR in T2DM patients.
Collapse
|
8
|
Minozzo BR, de Andrade EA, Vellosa JCR, Lipinski LC, Fernandes D, Nardi GM, Rodrigues RP, Kitagawa RR, Girard C, Demougeot C, Beltrame FL. Polyphenolic compounds of Euphorbia umbellata (Pax) Bruyns (Euphorbiaceae) improved endothelial dysfunction through arginase inhibition. Phytother Res 2021; 35:2557-2567. [PMID: 33350522 DOI: 10.1002/ptr.6986] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 12/04/2020] [Accepted: 12/04/2020] [Indexed: 11/30/2023]
Abstract
Euphorbia umbellata is used for its anti-inflammatory properties; however, there are limited data available regarding its effects on vascular function. Its bark is rich in polyphenolic compounds, which potentially improve endothelial dysfunction (ED). This study proposes to investigate the effects of E. umbellata bark extracts and its polyphenolic compounds on arginase (ARG) activity and nitric oxide (NO)-related targets. Chromatographic procedures were used for the chemical characterisation of the extracts. Furthermore, in silico (molecular docking), in vitro (ARG inhibition), in vivo (streptozotocin-induced hyperglycemia model), and ex vivo (l-arginine metabolism, vascular reactivity, western blot, and biochemical) techniques were carried out. Quercetin, gallic acid, and ellagic acid were identified in the extracts. In silico screening predicted that gallic acid and quercetin would have the most promising interactions with ARG -identified cavities. This was confirmed in vitro as both compounds had a direct inhibitory effect on ARG, as was the case regarding the extracts. Oral treatment preserved endothelium-dependent vasodilation through ARG inhibition together with an increase in l-arginine bioavailability and endothelial NO synthase expression. Biochemical parameters determined the lack of toxicity for sub-chronic treatment. E. umbellata bark extracts and its compounds can contribute to ED treatment, at least partly, through the inhibition of vascular ARG.
Collapse
Affiliation(s)
- Bruno Rodrigo Minozzo
- Department of Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, Brazil
| | - Evelyn Assis de Andrade
- Department of Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, Brazil
| | | | | | - Daniel Fernandes
- Department of Pharmacology, Federal University of Santa Catarina, Florianópolis, Brazil
| | - Geisson Marcos Nardi
- Department of Morphology, Federal University of Santa Catarina, Florianópolis, Brazil
| | | | | | - Corine Girard
- PEPITE EA 4267, University Bourgogne Franche-Comté, Besançon, France
| | - Céline Demougeot
- PEPITE EA 4267, University Bourgogne Franche-Comté, Besançon, France
| | - Flávio Luís Beltrame
- Department of Pharmaceutical Sciences, State University of Ponta Grossa, Ponta Grossa, Brazil
| |
Collapse
|
9
|
Zahorán S, Szántó PR, Bódi N, Bagyánszki M, Maléth J, Hegyi P, Sári T, Hermesz E. Sustained Maternal Smoking Triggers Endothelial-Mediated Oxidative Stress in the Umbilical Cord Vessels, Resulting in Vascular Dysfunction. Antioxidants (Basel) 2021; 10:antiox10040583. [PMID: 33918732 PMCID: PMC8069726 DOI: 10.3390/antiox10040583] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/15/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023] Open
Abstract
Nitric oxide (NO) bioavailability is fundamental in the regulation of redox balance and functionality of the endothelium, especially in the case of the umbilical cord (UC), which has no innervation. The analysis of UC vessel-related complications could serve as a useful tool in the understanding of the pathophysiological mechanisms leading to neonatal cardiovascular disorders. Therefore, the aim of this study was to characterize the mechanisms that rule the severity of prenatal endothelial dysfunction, induced by the long-term effect of maternal smoking. Our analysis describes the initiation and the consequences of endothelial nitric oxide synthase (NOS3) deactivation, along with the up-regulation of possible compensatory pathways, using structural, molecular and biochemical approaches. This study was carried out on both the UC arteries and veins originated from neonates born to non-smoking and heavy-smoking mothers. The alterations stimulated by maternal smoking are vessel-specific and proportional to the level of exposure to harmful materials passed through the placenta. Typically, in the primarily exposed veins, an increased formation of reactive oxygen species and an up-regulation of the highly-efficient NOS2-NO producing pathway were detected. Despite all the extensive structural and functional damages, the ex vivo heat and cadmium ion-treated UC vein pieces still support the potential for stress response.
Collapse
Affiliation(s)
- Szabolcs Zahorán
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (S.Z.); (P.R.S.)
| | - Péter R. Szántó
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (S.Z.); (P.R.S.)
| | - Nikolett Bódi
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (N.B.); (M.B.)
| | - Mária Bagyánszki
- Department of Physiology, Anatomy, and Neuroscience, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (N.B.); (M.B.)
| | - József Maléth
- First Department of Medicine, University of Szeged, H-6701 Szeged, Hungary;
- HAS-USZ Momentum Epithel Cell Signalling and Secretion Research Group, H-6701 Szeged, Hungary
| | - Péter Hegyi
- Institute for Translational Medicine, Medical School, University of Pécs, H-7601 Pécs, Hungary;
| | - Tamás Sári
- Department of Obstetrics and Gynaecology, Faculty of Medicine, University of Szeged, H-6701 Szeged, Hungary;
| | - Edit Hermesz
- Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, H-6701 Szeged, Hungary; (S.Z.); (P.R.S.)
- Correspondence: ; Tel.: +36-(62)-544-887
| |
Collapse
|
10
|
Shatanawi A, Momani MS, Al-Aqtash R, Hamdan MH, Gharaibeh MN. L-Citrulline Supplementation Increases Plasma Nitric Oxide Levels and Reduces Arginase Activity in Patients With Type 2 Diabetes. Front Pharmacol 2020; 11:584669. [PMID: 33414716 PMCID: PMC7783447 DOI: 10.3389/fphar.2020.584669] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/05/2020] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is becoming a major contributor to cardiovascular disease. One of the early signs of T2DM associated cardiovascular events is the development of vascular dysfunction. This dysfunction has been implicated in increasing the morbidity and mortality of T2DM patients. One of the important characteristics of vascular dysfunction is the impaired ability of endothelial cells to produce nitric oxide (NO). Additionally, decreases in the availability of NO is also a major contributor of this pathology. NO is produced by the activity of endothelial NO synthase (eNOS) on its substrate, L-arginine. Reduced availability of L-arginine to eNOS has been implicated in vascular dysfunction in diabetes. Arginase, which metabolizes L-arginine to urea and ornithine, competes directly with NOS for L-arginine. Hence, increases in arginase activity can decrease arginine levels, reducing its availability to eNOS and decreasing NO production. Diabetes has been linked to elevated arginase and associated vascular endothelial dysfunction. We aimed to determine levels of plasma NO and arginase activity in (T2DM) patients and the effects of L-citrulline supplementation, a natural arginase inhibitor, on inhibiting arginase activity in these patients. Levels of arginase correlated with HbA1c levels in diabetic patients. Twenty-five patients received L-citrulline supplements (2000 mg/day) for 1 month. Arginase activity decreased by 21% in T2DM patients after taking L-citrulline supplements. Additionally, plasma NO levels increased by 38%. There was a modest improvement on H1Ac levels in these patients, though not statistically significant. The effect of L-citrulline on arginase activity was also studied in bovine aortic endothelial cells (BAECs) grown in high glucose (HG) conditions. HG (25 mM, 72 h) caused a 2-fold increase in arginase activity in BAECs and decreased NO production by 30%. L-citrulline (2.5 mM) completely prevented the increase in arginase activity and restored NO production levels. These data indicate that L-citrulline can have therapeutic benefits in diabetic patients through increasing NO levels and thus maintaining vascular function possibly through an arginase inhibition related pathway.
Collapse
Affiliation(s)
- Alia Shatanawi
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Munther S Momani
- Department of Internal Medicine, School of Medicine, The University of Jordan, Amman, Jordan
| | - Ruaa Al-Aqtash
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| | - Mohammad H Hamdan
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan.,Department of Neurosurgery, Saarland University Hospital, Homburg, Germany
| | - Munir N Gharaibeh
- Department of Pharmacology, School of Medicine, The University of Jordan, Amman, Jordan
| |
Collapse
|
11
|
A Protein/Lipid Preload Attenuates Glucose-Induced Endothelial Dysfunction in Individuals with Abnormal Glucose Tolerance. Nutrients 2020; 12:nu12072053. [PMID: 32664350 PMCID: PMC7400934 DOI: 10.3390/nu12072053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/24/2020] [Accepted: 07/08/2020] [Indexed: 12/24/2022] Open
Abstract
Postprandial hyperglycemia interferes with vascular reactivity and is a strong predictor of cardiovascular disease. Macronutrient preloads reduce postprandial hyperglycemia in subjects with impaired glucose tolerance (IGT) or type 2 diabetes (T2D), but the effect on endothelial function is unknown. Therefore, we examined whether a protein/lipid preload can attenuate postprandial endothelial dysfunction by lowering plasma glucose responses in subjects with IGT/T2D. Endothelial function was assessed by the reactive hyperemia index (RHI) at fasting, 60 min and 120 min during two 75 g oral glucose tolerance tests (OGTTs) preceded by either water or a macronutrient preload (i.e., egg and parmesan cheese) in 22 volunteers with IGT/T2D. Plasma glucose, insulin, glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic polypeptide (GIP), glucagon, free fatty acids, and amino acids were measured through each test. RHI negatively correlated with fasting plasma glucose. During the control OGTT, RHI decreased by 9% and its deterioration was associated with the rise in plasma glucose. The macronutrient preload attenuated the decline in RHI and markedly reduced postprandial glycemia. The beneficial effect of the macronutrient preload on RHI was proportional to the improvement in glucose tolerance and was associated with the increase in plasma GLP-1 and arginine levels. In conclusion, a protein/lipid macronutrient preload attenuates glucose-induced endothelial dysfunction in individuals with IGT/T2D by lowering plasma glucose excursions and by increasing GLP-1 and arginine levels, which are known regulators of the nitric oxide vasodilator system.
Collapse
|
12
|
Ichikawa S, Gohda T, Murakoshi M, Li Z, Adachi E, Koshida T, Suzuki Y. Aspartic acid supplementation ameliorates symptoms of diabetic kidney disease in mice. FEBS Open Bio 2020; 10:1122-1134. [PMID: 32301275 PMCID: PMC7262904 DOI: 10.1002/2211-5463.12862] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/31/2020] [Accepted: 04/13/2020] [Indexed: 12/23/2022] Open
Abstract
Diabetic kidney disease (DKD) is among the most common and serious complications of both type 1 and type 2 diabetes. In this study, we used KK/Ta‐Ins2Akita (KK‐Akita) mice as a model of DKD and KK/Ta (KK) mice as controls to identify novel factors related to the development/progression of DKD. Capillary electrophoresis coupled with mass spectrometry analysis revealed that circulating Asp (l‐aspartic acid) levels in diabetic KK‐Akita mice tend to be lower than those in control KK mice. Therefore, we evaluated the effect of Asp supplementation to prevent the progression of DKD in KK‐Akita mice. Mice were divided into three groups: (a) untreated KK mice (Control group), (b) untreated KK‐Akita mice (DKD group), and (c) treated (double‐volume Asp diet) KK‐Akita mice (Tx group). Kidney sections were stained with fluorescein isothiocyanate‐labeled lectins, wheat germ agglutinin (WGA), and anti‐endothelial nitric oxide synthase (eNOS) antibody for evaluation of endothelial surface layer (ESL) and NO synthesis. The mesangial area and glomerular size in the DKD group were significantly larger than those in the Control group; however, there was no significant difference in those between the DKD and Tx groups. Albuminuria, the ratio of foot process effacement, and thickness of glomerular basement membrane in the Tx group were significantly lower than those in the DKD group. Furthermore, the expression levels of glomerular WGA and microvascular eNOS in the Tx group improved significantly and approached the level in the Control group. In conclusion, the improvement of albuminuria in the Tx group may be caused by the reduction of oxidative stress in the kidneys, which may lead to the subsequent improvement of glomerular ESL.
Collapse
Affiliation(s)
- Saki Ichikawa
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Tomohito Gohda
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Maki Murakoshi
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Zi Li
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Eri Adachi
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Takeo Koshida
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Suzuki
- Department of Nephrology, Juntendo University Faculty of Medicine, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
13
|
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: 1.0] [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
|
14
|
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: 17] [Impact Index Per Article: 3.4] [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
|
15
|
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: 8.2] [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
|
16
|
Canè S, Bronte V. Detection and functional evaluation of arginase-1 isolated from human PMNs and murine MDSC. Methods Enzymol 2020; 632:193-213. [DOI: 10.1016/bs.mie.2019.07.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
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: 1.3] [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]
|
18
|
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: 2.3] [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
|
19
|
Role of Arginase 2 in Systemic Metabolic Activity and Adipose Tissue Fatty Acid Metabolism in Diet-Induced Obese Mice. Int J Mol Sci 2019; 20:ijms20061462. [PMID: 30909461 PMCID: PMC6472154 DOI: 10.3390/ijms20061462] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/14/2019] [Accepted: 03/19/2019] [Indexed: 12/23/2022] Open
Abstract
Visceral adipose tissue (VAT) inflammation and metabolic dysregulation are key components of obesity-induced metabolic disease. Upregulated arginase, a ureahydrolase enzyme with two isoforms (A1-cytosolic and A2-mitochondrial), is implicated in pathologies associated with obesity and diabetes. This study examined A2 involvement in obesity-associated metabolic and vascular disorders. WT and globally deleted A2(−/−) or A1(+/−) mice were fed either a high fat/high sucrose (HFHS) diet or normal diet (ND) for 16 weeks. Increases in body and VAT weight of HFHS-fed WT mice were abrogated in A2−/−, but not A1+/−, mice. Additionally, A2−/− HFHS-fed mice exhibited higher energy expenditure, lower blood glucose, and insulin levels compared to WT HFHS mice. VAT and adipocytes from WT HFHS fed mice showed greater A2 expression and adipocyte size and reduced expression of PGC-1α, PPAR-γ, and adiponectin. A2 deletion blunted these effects, increased levels of active AMPK-α, and upregulated genes involved in fatty acid metabolism. A2 deletion prevented HFHS-induced VAT collagen deposition and inflammation, which are involved in adipocyte metabolic dysfunction. Endothelium-dependent vasorelaxation, impaired by HFHS diet, was significantly preserved in A2−/− mice, but more prominently maintained in A1+/− mice. In summary, A2 is critically involved in HFHS-induced VAT inflammation and metabolic dysfunction.
Collapse
|
20
|
Shah SFA, Khan MJ, Iqbal T, Akram S, Waheed F, Satti HS, Rafiq MA, Hussain S. Arginase-1 Variants and the Risk of Familial Coronary Artery Disease in Subjects Originating from Pakistan. Genet Test Mol Biomarkers 2019; 23:32-38. [DOI: 10.1089/gtmb.2018.0227] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Syed Fawad Ali Shah
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | | | - Tahir Iqbal
- Department of Internal Medicine, Shifa College of Medicine, Shifa International Hospital, Islamabad, Pakistan
| | - Sumaira Akram
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Farah Waheed
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| | - Humayoon Shafique Satti
- Department of Hematology/Medicine, Armed Forces Bone Marrow Transplant Centre, Rawalpindi, Pakistan
| | | | - Sabir Hussain
- Department of Biosciences, COMSATS University Islamabad, Islamabad, Pakistan
| |
Collapse
|
21
|
Chandra S, Fulton DJR, Caldwell RB, Caldwell RW, Toque HA. Hyperglycemia-impaired aortic vasorelaxation mediated through arginase elevation: Role of stress kinase pathways. Eur J Pharmacol 2018; 844:26-37. [PMID: 30502342 DOI: 10.1016/j.ejphar.2018.11.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 11/15/2022]
Abstract
Diabetes-induced vascular endothelial dysfunction has been reported to involve hyperglycemia-induced increases in arginase activity. However, upstream mediators of this effect are not clear. Here, we have tested involvement of Rho kinase, ERK1/2 and p38 MAPK pathways in this process. Studies were performed with aortas isolated from wild type or hemizygous arginase 1 knockout (Arg1+/-) mice and bovine aortic endothelial cells exposed to high glucose (HG, 25 mmol/l) or normal glucose (NG, 5.5 mmol/l) conditions for different times. Effects of inhibitors of arginase, p38 MAPK, ERK1/2 or ROCK and ex vivo adenoviral delivery of active Arg1 and inactive (D128-Arg1) cDNA were also determined. Exposure in wild type aorta or endothelial cells to HG significantly increased arginase activity and Arg1 expression and impaired aortic relaxation. Transduction of wild type aorta with active Arg1 cDNA impaired vascular relaxation, whereas inactive Arg1 had no effect. The HG-induced vascular endothelial dysfunction was associated with increased phosphorylation (activation) of ERK1/2 and p38 MAPK. Pretreatment with inhibitors of ERK1/2, p38 MAPK, ROCK or arginase blocked HG-induced elevation of arginase activity and Arg1 expression and prevented the vascular dysfunction. Inhibition of ROCK blunted the HG-induced activation of ERK1/2 and p38 MAPK. In summary, activated ROCK and subsequent activation of ERK1/2 or p38 MAPK elevates arginase activity and Arg1 expression in hyperglycemic states. Targeting this pathway may provide an effective means for preventing diabetes/hyperglycemia-induced vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Surabhi Chandra
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Department of Biology, University of Nebraska-Kearney, Kearney, NE, USA.
| | - David J R Fulton
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Ruth B Caldwell
- Vascular Biology Center, Augusta University, Augusta, GA, USA; Cell Biology and Anatomy, Augusta University, Augusta, GA, USA; Veterans Administration Medical Center, Augusta, GA, USA
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Augusta University, Augusta, GA, USA; Vascular Biology Center, Augusta University, Augusta, GA, USA
| |
Collapse
|
22
|
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: 0.7] [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
|
23
|
Shah SFA, Iqbal T, Qamar R, Rafiq MA, Hussain S. ARG1Gene Polymorphisms and Their Association in Individuals with Essential Hypertension: A Case–Control Study. DNA Cell Biol 2018; 37:609-616. [DOI: 10.1089/dna.2018.4222] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Affiliation(s)
- Syed Fawad Ali Shah
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - Tahir Iqbal
- Department of Internal Medicine, Shifa College of Medicine, Shifa International Hospital, Islamabad, Pakistan
| | - Raheel Qamar
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - Muhammad Arshad Rafiq
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - Sabir Hussain
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| |
Collapse
|
24
|
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: 1.0] [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
|
25
|
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: 7.4] [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
|
26
|
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: 304] [Impact Index Per Article: 43.4] [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
|
27
|
Tavares G, Venturini G, Padilha K, Zatz R, Pereira AC, Thadhani RI, Rhee EP, Titan SMO. 1,5-Anhydroglucitol predicts CKD progression in macroalbuminuric diabetic kidney disease: results from non-targeted metabolomics. Metabolomics 2018; 14:39. [PMID: 30830377 DOI: 10.1007/s11306-018-1337-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/06/2018] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Metabolomics allows exploration of novel biomarkers and provides insights on metabolic pathways associated with disease. To date, metabolomics studies on CKD have been largely limited to Caucasian populations and have mostly examined surrogate end points. OBJECTIVE In this study, we evaluated the role of metabolites in predicting a primary outcome defined as dialysis need, doubling of serum creatinine or death in Brazilian macroalbuminuric DKD patients. METHODS Non-targeted metabolomics was performed on plasma from 56 DKD patients. Technical triplicates were done. Metabolites were identified using Agilent Fiehn GC/MS Metabolomics and NIST libraries (Agilent MassHunter Work-station Quantitative Analysis, version B.06.00). After data cleaning, 186 metabolites were left for analyses. RESULTS During a median follow-up time of 2.5 years, the PO occurred in 17 patients (30.3%). In non-parametric testing, 13 metabolites were associated with the PO. In univariate Cox regression, only 1,5-anhydroglucitol (HR 0.10; 95% CI 0.01-0.63, p = .01), norvaline and L-aspartic acid were associated with the PO. After adjustment for baseline renal function, 1,5-anhydroglucitol (HR 0.10; 95% CI 0.02-0.63, p = .01), norvaline (HR 0.01; 95% CI 0.001-0.4, p = .01) and aspartic acid (HR 0.12; 95% CI 0.02-0.64, p = .01) remained significantly and inversely associated with the PO. CONCLUSION Our results show that lower levels of 1,5-anhydroglucitol, norvaline and L-aspartic acid are associated with progression of macroalbuminuric DKD. While norvaline and L-aspartic acid point to interesting metabolic pathways, 1,5-anhydroglucitol is of particular interest since it has been previously shown to be associated with incident CKD. This inverse biomarker of hyperglycemia should be further explored as a new tool in DKD.
Collapse
Affiliation(s)
- Gesiane Tavares
- Nephrology Division, University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 255, São Paulo, SP, 05403-000, Brazil.
| | - Gabriela Venturini
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Kallyandra Padilha
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Roberto Zatz
- Nephrology Division, University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 255, São Paulo, SP, 05403-000, Brazil
| | - Alexandre C Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo Medical School, São Paulo, SP, Brazil
| | - Ravi I Thadhani
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Eugene P Rhee
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Silvia M O Titan
- Nephrology Division, University of São Paulo Medical School, Av Dr Enéas de Carvalho Aguiar, 255, São Paulo, SP, 05403-000, Brazil
| |
Collapse
|
28
|
Gong J, Du X, Li Z, Li X, Guo M, Lu J, Wang Y, Chen Z, Li C. 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: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
Affiliation(s)
- Jingjing Gong
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaoyan Du
- Department of Laboratory Animal, Capital Medical University, Beijing, China
| | - Zhenkun Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaohong Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Meng Guo
- Department of Laboratory Animal, Capital Medical University, Beijing, China
| | - Jing Lu
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Laboratory Animal, Capital Medical University, Beijing, China
| | - Zhenwen Chen
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Changlong Li
- School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
29
|
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: 8.4] [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
|
30
|
Vitak T, Yurkiv B, Wasser S, Nevo E, Sybirna N. Effect of medicinal mushrooms on blood cells under conditions of diabetes mellitus. World J Diabetes 2017; 8:187-201. [PMID: 28572880 PMCID: PMC5437617 DOI: 10.4239/wjd.v8.i5.187] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 02/13/2017] [Accepted: 03/12/2017] [Indexed: 02/05/2023] Open
Abstract
Diabetes mellitus (DM) is the third most common non-infectious disease leading to early disability and high mortality. Moreover, the number of patients is growing every year. The main symptom of DM is hyperglycemia. Increased levels of blood glucose activate polyol, hexosamine, and protein kinase metabolic pathways cause the intensification of non-enzymatic glycosylation and nitration of macromolecules. This, in turn, leads to the development of oxidative and nitrative stresses and secondary complications, such as different kinds of micro- and macroangiopathies. Metabolic disorders caused by insulin deficiency in diabetes significantly impede the functioning of a homeostasis system, which change the physical, biochemical, morphological, and functional properties of blood cells. As a result, the oxygen-transport function of red blood cells (RBCs), rheological properties of the blood, and functions of immunocompetent cells as well as the process of apoptosis are primarily affected. Modern pharmacotherapy focuses on the search for new preparations that aim to decrease blood glucose levels. Undesirable side effects and adverse reactions caused by synthetic medicines led to the search and investigation of new preparations of natural origin. Medicinal mushrooms play an important role among such new preparations. They are a source of a large number of high- and low-molecular compounds with pronounced biological effects. Our investigations show pronounced hypoglycemic and anti-anemic action of submerged cultivated mycelium powder of medicinal mushrooms Agaricus brasiliensis (A. brasiliensis) and Ganoderma lucidum (G. lucidum) on streptozotocin-induced DM in rats. Also, we showed that mycelium powders have membrane protective properties as evidenced by the redistribution of RBC populations towards the growth of full functional cell numbers. Normalization of parameters of leukocyte formula and suppression of apoptosis of white blood cells in diabetic rats treated with A. brasiliensis and G. lucidum mycelia indicates pronounced positive effects of these strains of mushrooms. Thus, the use of medicinal mushrooms for treatment of DM and in prevention development of its secondary complications might be a new effective approach of this disease's cure. This article is aimed at summarizing and analyzing the literature data and basic achievements concerning DM type 1 treatment using medicinal mushrooms and showing the results obtained in our research.
Collapse
|
31
|
Lamuchi-Deli N, Aberomand M, Babaahmadi-Rezaei H, Mohammadzadeh G. Effects of the Hydroalcoholic Extract of Zingiber officinale on Arginase I Activity and Expression in the Retina of Streptozotocin-Induced Diabetic Rats. Int J Endocrinol Metab 2017; 15:e42161. [PMID: 28835766 PMCID: PMC5555732 DOI: 10.5812/ijem.42161] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 01/23/2017] [Accepted: 02/01/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Emerging evidence suggests that an increased arginase activity is involved in vascular dysfunction in experimental animals. Zingiber officinale Roscoe, commonly known as ginger, has been widely used in the traditional medicine for treatment of diabetes. OBJECTIVES This study aimed at investigating the effects of the hydroalcoholic extract of Z. officinale on arginase I activity and expression in the retina of streptozotocin (STZ)-induced diabetic rats. METHODS In this experimental study, 16 male Wistar rats weighing 200 - 250 g were assessed. Diabetes was induced via a single intraperitoneal injection of STZ (60 mg/kg body weight). The rats were randomly allocated into four experimental groups. Untreated healthy and diabetic controls received 1.5 mL/kg distilled water. Treated diabetic rats received 200, and 400 mg/kg of the Z. officinale extract dissolved in distilled water (1.5 mL/kg). Body weight, blood glucose and insulin concentration were measured by standard methods. The arginase I activity and expression were determined by spectrophotometric and western blot analysis, respectively. RESULTS Our results showed that blood glucose concentration was significantly decreased in diabetic rats treated with the extract compared to untreated diabetic controls (P < 0.01). Treatment with 400 mg/kg of the extract reduced arginase I activity and expression (P < 0.05). A significant elevation in body weight was observed in diabetic rats treated with the extract. Serum insulin was significantly increased in diabetic rats treated with 400 mg/kg of the extract compared to diabetic controls (P < 0.05). CONCLUSIONS Our results suggest that the Z. officinale hydroalcoholic extract may potentially be a promising therapeutic option for treating diabetes-induced vascular disorders, possibly through reducing arginase I activity and expression in the retina.
Collapse
Affiliation(s)
- Nasrin Lamuchi-Deli
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Mohammad Aberomand
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Hossein Babaahmadi-Rezaei
- Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
| | - Ghorban Mohammadzadeh
- Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran
- Corresponding author: Ghorban Mohammadzadeh, Hyperlipidemia Research Center, Department of Clinical Biochemistry, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, IR Iran. Tel: +98-09113436812, Fax: +98-6133332036, E-mail:
| |
Collapse
|
32
|
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: 86.5] [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
|
33
|
Vanhoutte PM, Shimokawa H, Feletou M, Tang EHC. Endothelial dysfunction and vascular disease - a 30th anniversary update. Acta Physiol (Oxf) 2017; 219:22-96. [PMID: 26706498 DOI: 10.1111/apha.12646] [Citation(s) in RCA: 620] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/27/2015] [Accepted: 12/17/2015] [Indexed: 02/06/2023]
Abstract
The endothelium can evoke relaxations of the underlying vascular smooth muscle, by releasing vasodilator substances. The best-characterized endothelium-derived relaxing factor (EDRF) is nitric oxide (NO) which activates soluble guanylyl cyclase in the vascular smooth muscle cells, with the production of cyclic guanosine monophosphate (cGMP) initiating relaxation. The endothelial cells also evoke hyperpolarization of the cell membrane of vascular smooth muscle (endothelium-dependent hyperpolarizations, EDH-mediated responses). As regards the latter, hydrogen peroxide (H2 O2 ) now appears to play a dominant role. Endothelium-dependent relaxations involve both pertussis toxin-sensitive Gi (e.g. responses to α2 -adrenergic agonists, serotonin, and thrombin) and pertussis toxin-insensitive Gq (e.g. adenosine diphosphate and bradykinin) coupling proteins. New stimulators (e.g. insulin, adiponectin) of the release of EDRFs have emerged. In recent years, evidence has also accumulated, confirming that the release of NO by the endothelial cell can chronically be upregulated (e.g. by oestrogens, exercise and dietary factors) and downregulated (e.g. oxidative stress, smoking, pollution and oxidized low-density lipoproteins) and that it is reduced with ageing and in the course of vascular disease (e.g. diabetes and hypertension). Arteries covered with regenerated endothelium (e.g. following angioplasty) selectively lose the pertussis toxin-sensitive pathway for NO release which favours vasospasm, thrombosis, penetration of macrophages, cellular growth and the inflammatory reaction leading to atherosclerosis. In addition to the release of NO (and EDH, in particular those due to H2 O2 ), endothelial cells also can evoke contraction of the underlying vascular smooth muscle cells by releasing endothelium-derived contracting factors. Recent evidence confirms that most endothelium-dependent acute increases in contractile force are due to the formation of vasoconstrictor prostanoids (endoperoxides and prostacyclin) which activate TP receptors of the vascular smooth muscle cells and that prostacyclin plays a key role in such responses. Endothelium-dependent contractions are exacerbated when the production of nitric oxide is impaired (e.g. by oxidative stress, ageing, spontaneous hypertension and diabetes). They contribute to the blunting of endothelium-dependent vasodilatations in aged subjects and essential hypertensive and diabetic patients. In addition, recent data confirm that the release of endothelin-1 can contribute to endothelial dysfunction and that the peptide appears to be an important contributor to vascular dysfunction. Finally, it has become clear that nitric oxide itself, under certain conditions (e.g. hypoxia), can cause biased activation of soluble guanylyl cyclase leading to the production of cyclic inosine monophosphate (cIMP) rather than cGMP and hence causes contraction rather than relaxation of the underlying vascular smooth muscle.
Collapse
Affiliation(s)
- P. M. Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| | - H. Shimokawa
- Department of Cardiovascular Medicine; Tohoku University; Sendai Japan
| | - M. Feletou
- Department of Cardiovascular Research; Institut de Recherches Servier; Suresnes France
| | - E. H. C. Tang
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
- School of Biomedical Sciences; Li Ka Shing Faculty of Medicine; The University of Hong Kong; Hong Kong City Hong Kong
| |
Collapse
|
34
|
Arginase 2 promotes neurovascular degeneration during ischemia/reperfusion injury. Cell Death Dis 2016; 7:e2483. [PMID: 27882947 PMCID: PMC5260867 DOI: 10.1038/cddis.2016.295] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/18/2016] [Accepted: 08/17/2016] [Indexed: 02/07/2023]
Abstract
Retinal ischemia is a major cause of visual impairment and blindness and is involved in various disorders including diabetic retinopathy, glaucoma, optic neuropathies and retinopathy of prematurity. Neurovascular degeneration is a common feature of these pathologies. Our lab has previously reported that the ureahydrolase arginase 2 (A2) is involved in ischemic retinopathies. Here, we are introducing A2 as a therapeutic target to prevent neurovascular injury after retinal ischemia/reperfusion (I/R) insult. Studies were performed with mice lacking both copies of A2 (A2−/−) and wild-type (WT) controls (C57BL6J). I/R insult was conducted on the right eye and the left eye was used as control. Retinas were collected for analysis at different times (3 h–4 week after injury). Neuronal and microvascular degeneration were evaluated using NeuN staining and vascular digests, respectively. Glial activation was evaluated by glial fibrillary acidic protein expression. Necrotic cell death was studied by propidium iodide labeling and western blot for RIP-3. Arginase expression was determined by western blot and quantitative RT-PCR. Retinal function was determined by electroretinography (ERG). A2 mRNA and protein levels were increased in WT I/R. A2 deletion significantly reduced ganglion cell loss and microvascular degeneration and preserved retinal morphology after I/R. Glial activation, reactive oxygen species formation and cell death by necroptosis were significantly reduced by A2 deletion. ERG showed improved positive scotopic threshold response with A2 deletion. This study shows for the first time that neurovascular injury after retinal I/R is mediated through increased expression of A2. Deletion of A2 was found to be beneficial in reducing neurovascular degeneration after I/R.
Collapse
|
35
|
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: 7.6] [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
|
36
|
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: 6.2] [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
|
37
|
Bhatta A, Sangani R, Kolhe R, Toque HA, Cain M, Wong A, Howie N, Shinde R, Elsalanty M, Yao L, Chutkan N, Hunter M, Caldwell RB, Isales C, Caldwell RW, Fulzele S. Deregulation of arginase induces bone complications in high-fat/high-sucrose diet diabetic mouse model. Mol Cell Endocrinol 2016; 422:211-220. [PMID: 26704078 PMCID: PMC4824063 DOI: 10.1016/j.mce.2015.12.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 12/03/2015] [Accepted: 12/04/2015] [Indexed: 01/21/2023]
Abstract
A balanced diet is crucial for healthy development and prevention of musculoskeletal related diseases. Diets high in fat content are known to cause obesity, diabetes and a number of other disease states. Our group and others have previously reported that activity of the urea cycle enzyme arginase is involved in diabetes-induced dysregulation of vascular function due to decreases in nitric oxide formation. We hypothesized that diabetes may also elevate arginase activity in bone and bone marrow, which could lead to bone-related complications. To test this we determined the effects of diabetes on expression and activity of arginase, in bone and bone marrow stromal cells (BMSCs). We demonstrated that arginase 1 is abundantly present in the bone and BMSCs. We also demonstrated that arginase activity and expression in bone and bone marrow is up-regulated in models of diabetes induced by HFHS diet and streptozotocin (STZ). HFHS diet down-regulated expression of healthy bone metabolism markers (BMP2, COL-1, ALP, and RUNX2) and reduced bone mineral density, bone volume and trabecular thickness. However, treatment with an arginase inhibitor (ABH) prevented these bone-related complications of diabetes. In-vitro study of BMSCs showed that high glucose treatment increased arginase activity and decreased nitric oxide production. These effects were reversed by treatment with an arginase inhibitor (ABH). Our study provides evidence that deregulation of l-arginine metabolism plays a vital role in HFHS diet-induced diabetic complications and that these complications can be prevented by treatment with arginase inhibitors. The modulation of l-arginine metabolism in disease could offer a novel therapeutic approach for osteoporosis and other musculoskeletal related diseases.
Collapse
Affiliation(s)
- Anil Bhatta
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Rajnikumar Sangani
- Departments of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Ravindra Kolhe
- Departments of Pathology, Georgia Regents University, Augusta, GA 30912, USA
| | - Haroldo A Toque
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | - Michael Cain
- Departments of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Abby Wong
- Departments of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Nicole Howie
- School of Dentistry, Georgia Regents University, Augusta, GA 30912, Augusta, GA 30912, USA
| | - Rahul Shinde
- Departments of Pathology, Georgia Regents University, Augusta, GA 30912, USA
| | - Mohammed Elsalanty
- School of Dentistry, Georgia Regents University, Augusta, GA 30912, Augusta, GA 30912, USA
| | - Lin Yao
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA
| | | | - Monty Hunter
- Departments of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA
| | - Ruth B Caldwell
- Cell Biology and Anatomy and Vascular Biology Center, Georgia Regents University; Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Carlos Isales
- Departments of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA
| | - R William Caldwell
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, GA 30912, USA.
| | - Sadanand Fulzele
- Departments of Orthopaedic Surgery, Georgia Regents University, Augusta, GA 30912, USA; Institute of Regenerative and Reparative Medicine, Georgia Regents University, Augusta, GA 30912, USA.
| |
Collapse
|
38
|
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: 3.0] [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
|
39
|
Zhang Y, Chen Q, Sun Z, Han J, Wang L, Zheng L. Impaired capsaicin-induced relaxation in diabetic mesenteric arteries. J Diabetes Complications 2015; 29:747-54. [PMID: 26055306 DOI: 10.1016/j.jdiacomp.2015.05.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Revised: 05/01/2015] [Accepted: 05/10/2015] [Indexed: 12/19/2022]
Abstract
AIMS To investigate capsaicin-induced vasodilation in the diabetic mesenteric arteries. METHODS A diabetic rat model was established by intraperitoneal injection of streptozotocin after a 12-h fast. At 12 weeks post-injection, the third branch of the mesenteric artery was dissected out and prepared for vascular reactivity assessment. Capsaicin, capsazepine, N(ω)-nitro-L-arginine methyl ester hydrochloride (L-NAME), sodium nitroprusside (SNP), calcitonin gene-related peptide 8-37 (CGRP8-37), calcitonin gene-related peptide (CGRP), and substance P (SP) were added to investigate subsequent alterations in vascular activity. Plasma and physiological salt solution (PSS) levels of CGRP and SP were measured using radioimmunoassay. The expression of transient receptor potential vanilloid 1 (TRPV1) and phospho-eNOS were determined using Western blot analysis, and nitric oxide (NO) production was measured using a fluorescence probe. RESULTS The dilation effect of capsaicin was weaker under the diabetic than control conditions. Capsazepine, L-NAME, and CGRP8-37 attenuated capsaicin-induced vasorelaxation significantly in the diabetic vascular rings. Exogenous CGRP elicited dose-dependent vasodilation in the control arteries, whereas the dilation effect was reduced under diabetic conditions. Plasma and PSS CGRP levels were attenuated and mesenteric artery TRPV1 expression was decreased in the diabetic rats. Phospho-eNOS levels were augmented, and NO production increased following the administration of capsaicin. CONCLUSIONS Decreased expression of TRPV1 and associated neuropeptide release contributed to the impaired capsaicin-induced vasodilation in diabetic mesenteric arteries. Furthermore, an endothelium-dependent NO-related pathway was involved in capsaicin-induced vasodilation.
Collapse
Affiliation(s)
- Yuanyuan Zhang
- Department of Cardiovascular Sciences, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Qiang Chen
- Department of Cardiovascular Sciences, Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu 322000, China
| | - Zewei Sun
- Department of Cardiovascular Sciences, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Jie Han
- Department of Cardiovascular Sciences, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Lihong Wang
- Department of Cardiovascular Sciences, Zhejiang Provincial People's Hospital, Hangzhou 310014, China
| | - Liangrong Zheng
- Department of Cardiovascular Sciences, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
40
|
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: 221] [Impact Index Per Article: 22.1] [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
|
41
|
Bhatta A, Yao L, Toque HA, Shatanawi A, Xu Z, Caldwell RB, Caldwell RW. Angiotensin II-induced arterial thickening, fibrosis and stiffening involves elevated arginase function. PLoS One 2015; 10:e0121727. [PMID: 25807386 PMCID: PMC4373900 DOI: 10.1371/journal.pone.0121727] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 02/03/2015] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Arterial stiffness (AS) is an independent risk factor for cardiovascular morbidity/mortality. Smooth muscle cell (SMC) proliferation and increased collagen synthesis are key features in development of AS. Arginase (ARG), an enzyme implicated in many cardiovascular diseases, can compete with nitric oxide (NO) synthase for their common substrate, L-arginine. Increased arginase can also provide ornithine for synthesis of polyamines via ornithine decarboxylase (ODC) and proline/collagen via ornithine aminotransferase (OAT), leading to vascular cell proliferation and collagen formation, respectively. We hypothesized that elevated arginase activity is involved in Ang II-induced arterial thickening, fibrosis, and stiffness and that limiting its activity can prevent these changes. METHODS AND RESULTS We tested this by studies in mice lacking one copy of the ARG1 gene that were treated with angiotensin II (Ang II, 4 weeks). Studies were also performed in rat aortic Ang II-treated SMC. In WT mice treated with Ang II, we observed aortic stiffening (pulse wave velocity) and aortic and coronary fibrosis and thickening that were associated with increases in ARG1 and ODC expression/activity, proliferating cell nuclear antigen, hydroxyproline levels, and collagen 1 protein expression. ARG1 deletion prevented each of these alterations. Furthermore, exposure of SMC to Ang II (1 μM, 48 hrs) increased ARG1 expression, ARG activity, ODC mRNA and activity, cell proliferation, collagen 1 protein expression and hydroxyproline content. Treatment with ABH prevented these changes. CONCLUSION Arginase 1 is crucially involved in Ang II-induced SMC proliferation and arterial fibrosis and stiffness and represents a promising therapeutic target.
Collapse
Affiliation(s)
- Anil Bhatta
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Lin Yao
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Haroldo A. Toque
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Alia Shatanawi
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| | - Zhimin Xu
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - Ruth B. Caldwell
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
| | - R. William Caldwell
- Department of Pharmacology and Toxicology, Georgia Regents University, Augusta, Georgia, United States of America
| |
Collapse
|
42
|
RhoA/mDia-1/profilin-1 signaling targets microvascular endothelial dysfunction in diabetic retinopathy. Graefes Arch Clin Exp Ophthalmol 2015; 253:669-80. [PMID: 25791356 DOI: 10.1007/s00417-015-2985-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 02/21/2015] [Accepted: 03/02/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Diabetic retinopathy (DR) is a major cause of blindness in the working-age populations of developed countries, and effective treatments and prevention measures have long been the foci of study. Patients with DR invariably demonstrate impairments of the retinal microvascular endothelium. Many observational and preclinical studies have shown that angiogenesis and apoptosis play crucial roles in the pathogenesis of DR. Increasing evidence suggests that in DR, the small guanosine-5'-triphosphate-binding protein RhoA activates its downstream targets mammalian Diaphanous homolog 1 (mDia-1) and profilin-1, thus affecting important cellular functions, including cell morphology, motility, secretion, proliferation, and gene expression. However, the specific underlying mechanism of disease remains unclear. CONCLUSION This review focuses on the RhoA/mDia-1/profilin-1 signaling pathway that specifically triggers endothelial dysfunction in diabetic patients. Recently, RhoA and profilin-1 signaling has attracted a great deal of attention in the context of diabetes-related research. However, the precise molecular mechanism by which the RhoA/mDia-1/profilin-1 pathway is involved in progression of microvascular endothelial dysfunction (MVED) during DR has not been determined. This review briefly describes each feature of the cascade before exploring the most recent findings on how the pathway may trigger endothelial dysfunction in DR. When the underlying mechanisms are understood, novel therapies seeking to restore the endothelial homeostasis comprised in DR will become possible.
Collapse
|
43
|
Zandifar A, Seifabadi S, Zandifar E, Beheshti SS, Aslani A, Javanmard SH. Comparison of the effect of topical versus systemic L-arginine on wound healing in acute incisional diabetic rat model. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2015; 20:233-8. [PMID: 26109968 PMCID: PMC4468226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2014] [Revised: 02/15/2015] [Accepted: 02/25/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Diabetes is associated with endothelial dysfunction and impaired wound healing. The amino acid L-arginine is the only substrate for nitric oxide (NO) synthesis. The purpose of this study was to compare the topical versus systemic L-arginine treatment on total nitrite (NOx) and vascular endothelial growth factor (VEGF) concentrations in wound fluid and rate of wound healing in an acute incisional diabetic wound model. MATERIALS AND METHODS A total of 56 Sprague-Dawley rats were used of which 32 were rendered diabetic. Animals underwent a dorsal skin incision. Dm-sys-arg group (N = 8, diabetic) and Norm-sys-arg group (N = 8, normoglycemic) were gavaged with L-arginine. Dm-sys-control group (N = 8, diabetic) and Norm-sys-control group (N = 8, normoglycemic) were gavaged with water. Dm-top-arg group (N = 8, diabetic) and norm-top-arg group (N = 8, normoglycemic) received topical L-arginine gel. Dm-top-control group (N = 8, diabetic) received gel vehicle. On the day 5 the amount of NOx in wound fluid was measured by Griess reaction. VEGF/total protein in wound fluids was also measured on day 5 using enzyme-linked immunosorbent assay. All wound tissue specimens were fixed and stained to be evaluated for rate of healing. Data were analyzed using SPSS software (version 18.0, Chicago, IL, USA) through One-way analysis of variance test and Tukey's post-hoc. RESULTS In dm-sys-arg group, the level of NOx on day 5 was significantly more than dm-top-arg group (P < 0.05). VEGF content in L-arginine treated groups were significantly more than controls (P < 0.05). Rate of diabetic wound healing in dm-sys-arg group was significantly more than dm-top-arg group. CONCLUSION Systemic L-arginine is more efficient than topical L-arginine in wound healing. This process is mediated at least in part, by increasing VEGF and NO in the wound fluid.
Collapse
Affiliation(s)
- Alireza Zandifar
- Physiology Research Center and Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran,Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sima Seifabadi
- Physiology Research Center and Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran,Pharmacy Student Research Center, Isfahan University of Medical Sciences, Isfahan,Department of Pharmaceutics and Novel Drug Delivery Systems Research Center, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ehsan Zandifar
- Physiology Research Center and Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran,Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sajedeh Sohrabi Beheshti
- Physiology Research Center and Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran,Medical Student Research Center, Isfahan University of Medical Sciences, Isfahan, Iran,Department of Physiology, Isfahan Payame Noor University, Isfahan, Iran
| | - Abolfazl Aslani
- Department of Pharmaceutics and Novel Drug Delivery Systems Research Center, School of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Physiology Research Center and Department of Physiology, Isfahan University of Medical Sciences, Isfahan, Iran,Address for correspondence: Dr. Shaghayegh Haghjooy Javanmard, Physiology Research Center, Isfahan University of Medical Sciences and Health Services, Hezar-Jerib St., Isfahan, Iran. E-mail: ,
| |
Collapse
|
44
|
Veeranki S, Tyagi SC. Role of hydrogen sulfide in skeletal muscle biology and metabolism. Nitric Oxide 2014; 46:66-71. [PMID: 25461301 DOI: 10.1016/j.niox.2014.11.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2014] [Revised: 11/12/2014] [Accepted: 11/21/2014] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide (H2S) is a novel endogenous gaseous signal transducer (gasotransmitter). Its emerging role in multiple facets of inter- and intra-cellular signaling as a metabolic, inflammatory, neuro and vascular modulator has been increasingly realized. Although H2S is known for its effects as an anti-hypertensive, anti-inflammatory and anti-oxidant molecule, the relevance of these effects in skeletal muscle biology during health and during metabolic syndromes is unclear. H2S has been implicated in vascular relaxation and vessel tone enhancement, which might lead to mitigation of vascular complications caused by the metabolic syndromes. Metabolic complications may also lead to mitochondrial remodeling by interfering with fusion and fission, therefore, leading to mitochondrial mitophagy and skeletal muscle myopathy. Mitochondrial protection by H2S enhancing treatments may mitigate deterioration of muscle function during metabolic syndromes. In addition, H2S might upregulate uncoupling proteins and might also cause browning of white fat, resulting in suppression of imbalanced cytokine signaling caused by abnormal fat accumulation. Likewise, as a source for H(+) ions, it has the potential to augment anaerobic ATP synthesis. However, there is a need for studies to test these putative H2S benefits in different patho-physiological scenarios before its full-fledged usage as a therapeutic molecule. The present review highlights current knowledge with regard to exogenous and endogenous H2S roles in skeletal muscle biology, metabolism, exercise physiology and related metabolic disorders, such as diabetes and obesity, and also provides future directions.
Collapse
Affiliation(s)
- Sudhakar Veeranki
- Department of Physiology & Biophysics, University of Louisville, Louisville, KY 40202, USA.
| | - Suresh C Tyagi
- Department of Physiology & Biophysics, University of Louisville, Louisville, KY 40202, USA
| |
Collapse
|
45
|
Suwanpradid J, Rojas M, Behzadian MA, Caldwell RW, Caldwell RB. Arginase 2 deficiency prevents oxidative stress and limits hyperoxia-induced retinal vascular degeneration. PLoS One 2014; 9:e110604. [PMID: 25375125 PMCID: PMC4222858 DOI: 10.1371/journal.pone.0110604] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Accepted: 09/16/2014] [Indexed: 01/02/2023] Open
Abstract
Background Hyperoxia exposure of premature infants causes obliteration of the immature retinal microvessels, leading to a condition of proliferative vitreoretinal neovascularization termed retinopathy of prematurity (ROP). Previous work has demonstrated that the hyperoxia-induced vascular injury is mediated by dysfunction of endothelial nitric oxide synthase resulting in peroxynitrite formation. This study was undertaken to determine the involvement of the ureahydrolase enzyme arginase in this pathology. Methods and Findings Studies were performed using hyperoxia-treated bovine retinal endothelial cells (BRE) and mice with oxygen-induced retinopathy (OIR) as experimental models of ROP. Treatment with the specific arginase inhibitor 2(S)-amino-6-boronohexanoic acid (ABH) prevented hyperoxia-induced apoptosis of BRE cells and reduced vaso-obliteration in the OIR model. Furthermore, deletion of the arginase 2 gene protected against hyperoxia-induced vaso-obliteration, enhanced physiological vascular repair, and reduced retinal neovascularization in the OIR model. Additional deletion of one copy of arginase 1 did not improve the vascular pathology. Analyses of peroxynitrite by quantitation of its biomarker nitrotyrosine, superoxide by dihydroethidium imaging and NO formation by diaminofluoroscein imaging showed that the protective actions of arginase 2 deletion were associated with blockade of superoxide and peroxynitrite formation and normalization of NOS activity. Conclusions Our data demonstrate the involvement of arginase activity and arginase 2 expression in hyperoxia-induced vascular injury. Arginase 2 deletion prevents hyperoxia-induced retinal vascular injury by preventing NOS uncoupling resulting in decreased reactive oxygen species formation and increased nitric oxide bioavailability.
Collapse
Affiliation(s)
- Jutamas Suwanpradid
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
| | - Modesto Rojas
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Pharmacology and Toxicology, Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, Georgia, United States of America
| | - M. Ali Behzadian
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
| | - R. William Caldwell
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
- Department of Pharmacology and Toxicology, Department of Cellular Biology & Anatomy, Georgia Regents University, Augusta, Georgia, United States of America
| | - Ruth B. Caldwell
- Vascular Biology Center, Georgia Regents University, Augusta, Georgia, United States of America
- Culver Vision Discovery Institute, Georgia Regents University, Augusta, Georgia, United States of America
- Charlie Norwood VA Medical Center, Augusta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
46
|
Chantler PD, Frisbee JC. Arterial function in cardio-metabolic diseases: from the microcirculation to the large conduits. Prog Cardiovasc Dis 2014; 57:489-96. [PMID: 25220256 DOI: 10.1016/j.pcad.2014.09.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
The metabolic syndrome (MetS) is characterized as a constellation of metabolic risk factors such as obesity, hypertension, dyslipidemia, and hyperglycemia that co-occur within a given individual. This consultation of risk factors exposes MetS to a 3-fold increased risk of cardiovascular disease and an even higher risk of developing type 2 diabetes compared to healthy individuals. The pathophysiological mechanisms underlying this increased cardiovascular risk are incompletely understood but likely include alterations to macro- and micro-vasculature. The vasculature plays an important role not only in delivery and adjusting the quantity of blood delivered to the tissues, but the dynamic changes in structure and compliance significantly alter the hemodynamic stress imposed on the heart and end-organs. This review will give an overview of the pathophysiological changes to the vasculature that accompany MetS in both human and animal models, as well as the possible mechanistic pathways.
Collapse
Affiliation(s)
- Paul D Chantler
- Division of Exercise Physiology, School of Medicine, West Virginia University, Morgantown, WV, USA; Center for Cardiovascular and Respiratory Sciences, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Jefferson C Frisbee
- Center for Cardiovascular and Respiratory Sciences, School of Medicine, West Virginia University, Morgantown, WV, USA; Department of Physiology and Pharmacology, School of Medicine, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
47
|
Toque HA, Caldwell RW. New approaches to the design and discovery of therapies to prevent erectile dysfunction. Expert Opin Drug Discov 2014; 9:1447-69. [DOI: 10.1517/17460441.2014.949234] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
48
|
Linderholm AL, Bratt JM, Schuster GU, Zeki AA, Kenyon NJ. Novel therapeutic strategies for adult obese asthmatics. Immunol Allergy Clin North Am 2014; 34:809-23. [PMID: 25282293 DOI: 10.1016/j.iac.2014.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Asthma is a complex syndrome that affects an estimated 26 million people in the United States but gaps exist in the recognition and management of asthmatic subgroups. This article proposes alternative approaches for future treatments of adult obese asthmatics who do not respond to standard controller therapies, drawing parallels between seemingly disparate therapeutics through their common signaling pathways. How metformin and statins can potentially improve airway inflammation is described and supplements are suggested. A move toward more targeted therapies for asthma subgroups is needed. These therapies address asthma and the comorbidities that accompany obesity and metabolic syndrome to provide the greatest therapeutic potential.
Collapse
Affiliation(s)
- Angela L Linderholm
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis, 4150 V Street, Suite 3100, Davis, CA, USA
| | - Jennifer M Bratt
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis, 4150 V Street, Suite 3100, Davis, CA, USA
| | - Gertrud U Schuster
- Nutrition Department, University of California, Davis, 430 West Health Sciences Drive, Davis, CA, USA; Immunity and Diseases Prevention Unit, Western Human Nutrition Research Center, United States Department of Agriculture (USDA), Agricultural Research Services (ARS), 430 West Health Sciences Drive, Davis, CA, USA
| | - Amir A Zeki
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis, 4150 V Street, Suite 3100, Davis, CA, USA
| | - Nicholas J Kenyon
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Internal Medicine, University of California, Davis, 4150 V Street, Suite 3100, Davis, CA, USA.
| |
Collapse
|
49
|
Persson P, Fasching A, Teerlink T, Hansell P, Palm F. l
-Citrulline, But Not
l
-Arginine, Prevents Diabetes Mellitus–Induced Glomerular Hyperfiltration and Proteinuria in Rat. Hypertension 2014; 64:323-9. [DOI: 10.1161/hypertensionaha.114.03519] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Diabetes mellitus–induced oxidative stress causes increased renal oxygen consumption and intrarenal tissue hypoxia. Nitric oxide is an important determinant of renal oxygen consumption and electrolyte transport efficiency. The present study investigates whether
l
-arginine or
l
-citrulline to promote nitric oxide production prevents the diabetes mellitus–induced kidney dysfunction. Glomerular filtration rate, renal blood flow, in vivo oxygen consumption, tissue oxygen tension, and proteinuria were investigated in control and streptozotocin-diabetic rats with and without chronic
l
-arginine or
l
-citrulline treatment for 3 weeks. Untreated and
l
-arginine–treated diabetic rats displayed increased glomerular filtration rate (2600±162 versus 1599±127 and 2290±171 versus 1739±138 µL/min per kidney), whereas
l
-citrulline prevented the increase (1227±126 versus 1375±88 µL/min per kidney). Filtration fraction was increased in untreated diabetic rats because of the increase in glomerular filtration rate but not in
l
-arginine– or
l
-citrulline–treated diabetic rats. Urinary protein excretion was increased in untreated and
l
-arginine–treated diabetic rats (142±25 versus 75±7 and 128±7 versus 89±7 µg/min per kidney) but not in diabetic rats administered
l
-citrulline (67±7 versus 61±5 µg/min per kidney). The diabetes mellitus–induced tissue hypoxia, because of elevated oxygen consumption, was unaltered by any of the treatments.
l
-citrulline administered to diabetic rats increases plasma
l
-arginine concentration, which prevents the diabetes mellitus–induced glomerular hyperfiltration, filtration fraction, and proteinuria, possibly by a vascular effect.
Collapse
Affiliation(s)
- Patrik Persson
- From the Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden (P.P., A.F., P.H., F.P.); Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands (T.T.); and Division of Drug Research, Department of Medical and Health Sciences (F.P.) and Center for Medical Image Science and Visualization (F.P.), Linköping University, Linköping, Sweden
| | - Angelica Fasching
- From the Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden (P.P., A.F., P.H., F.P.); Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands (T.T.); and Division of Drug Research, Department of Medical and Health Sciences (F.P.) and Center for Medical Image Science and Visualization (F.P.), Linköping University, Linköping, Sweden
| | - Tom Teerlink
- From the Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden (P.P., A.F., P.H., F.P.); Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands (T.T.); and Division of Drug Research, Department of Medical and Health Sciences (F.P.) and Center for Medical Image Science and Visualization (F.P.), Linköping University, Linköping, Sweden
| | - Peter Hansell
- From the Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden (P.P., A.F., P.H., F.P.); Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands (T.T.); and Division of Drug Research, Department of Medical and Health Sciences (F.P.) and Center for Medical Image Science and Visualization (F.P.), Linköping University, Linköping, Sweden
| | - Fredrik Palm
- From the Division of Integrative Physiology, Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden (P.P., A.F., P.H., F.P.); Department of Clinical Chemistry, VU University Medical Center, Amsterdam, The Netherlands (T.T.); and Division of Drug Research, Department of Medical and Health Sciences (F.P.) and Center for Medical Image Science and Visualization (F.P.), Linköping University, Linköping, Sweden
| |
Collapse
|
50
|
Sawada N, Liao JK. Rho/Rho-associated coiled-coil forming kinase pathway as therapeutic targets for statins in atherosclerosis. Antioxid Redox Signal 2014; 20:1251-67. [PMID: 23919640 PMCID: PMC3934442 DOI: 10.1089/ars.2013.5524] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE The 3-hydroxy-methylglutaryl coenzyme A reductase inhibitors or statins are important therapeutic agents for lowering serum cholesterol levels. However, recent studies suggest that statins may exert atheroprotective effects beyond cholesterol lowering. These so-called "pleiotropic effects" include effects of statins on vascular and inflammatory cells. Thus, it is important to understand whether other signaling pathways that are involved in atherosclerosis could be targets of statins, and if so, whether individuals with "overactivity" of these pathways could benefit from statin therapy, regardless of serum cholesterol level. RECENT ADVANCES Statins inhibit the synthesis of isoprenoids, which are important for the function of the Rho/Rho-associated coiled-coil containing kinase (ROCK) pathway. Indeed, recent studies suggest that inhibition of the Rho/ROCK pathway by statins could lead to improved endothelial function and decreased vascular inflammation and atherosclerosis. Thus, the Rho/ROCK pathway has emerged as an important target of statin therapy for reducing atherosclerosis and possibly cardiovascular disease. CRITICAL ISSUES Because atherosclerosis is both a lipid and an inflammatory disease, it is important to understand how inhibition of Rho/ROCK pathway could contribute to statins' antiatherosclerotic effects. FUTURE DIRECTIONS The role of ROCKs (ROCK1 and ROCK2) in endothelial, smooth muscle, and inflammatory cells needs to be determined in the context of atherogenesis. This could lead to the development of specific ROCK1 or ROCK2 inhibitors, which could have greater therapeutic benefits with less toxicity. Also, clinical trials will need to be performed to determine whether inhibition of ROCKs, with and without statins, could lead to further reduction in atherosclerosis and cardiovascular disease.
Collapse
Affiliation(s)
- Naoki Sawada
- 1 GCOE Program and Department of Molecular Endocrinology and Metabolism, Tokyo Medical and Dental University , Tokyo, Japan
| | | |
Collapse
|