1
|
Jeon H, Lee D, Kim JY, Shim JJ, Lee JH. Limosilactobacillus reuteri HY7503 and Its Cellular Proteins Alleviate Endothelial Dysfunction by Increasing Nitric Oxide Production and Regulating Cell Adhesion Molecule Levels. Int J Mol Sci 2024; 25:11326. [PMID: 39457107 PMCID: PMC11509054 DOI: 10.3390/ijms252011326] [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: 10/02/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Endothelial dysfunction, which is marked by a reduction in nitric oxide (NO) production or an imbalance in relaxing and contracting factor levels, exacerbates atherosclerosis by promoting the production of cell adhesion molecules and cytokines. This study aimed to investigate the effects of Limosilactobacillus reuteri HY7503, a novel probiotic isolated from raw milk, on endothelial dysfunction. Five lactic acid bacterial strains were screened for their antioxidant, anti-inflammatory, and endothelium-protective properties; L. reuteri HY7503 had the most potent effect. In a mouse model of angiotensin II-induced endothelial dysfunction, L. reuteri HY7503 reduced vascular thickening (19.78%), increased serum NO levels (226.70%), upregulated endothelial NO synthase (eNOS) expression in the aortic tissue, and decreased levels of cell adhesion molecules (intercellular adhesion molecule-1 [ICAM-1] and vascular cell adhesion molecule-1 [VCAM-1]) and serum cytokines (tumor necrosis factor-alpha [TNF-α] and interleukin-6 [IL-6]). In TNF-α-treated human umbilical vein endothelial cells (HUVECs), L. reuteri HY7503 enhanced NO production and reduced cell adhesion molecule levels. In HUVECs, surface-layer proteins (SLPs) were more effective than extracellular vesicles (exosomes) in increasing NO production and decreasing cell adhesion molecule levels. These findings suggested that L. reuteri HY7503 may serve as a functional probiotic that alleviates endothelial dysfunction.
Collapse
Affiliation(s)
| | | | - Joo-Yun Kim
- R&BD Center, Hy Co., Ltd., 22 Giheungdanji-ro 24 Beon-gil, Giheung-gu, Yongin-si 17086, Gyeonggi-do, Republic of Korea; (H.J.); (D.L.); (J.-J.S.); (J.-H.L.)
| | | | | |
Collapse
|
2
|
Heuser SK, Li J, Pudewell S, LoBue A, Li Z, Cortese-Krott MM. Biochemistry, pharmacology and in vivo function of arginases. Pharmacol Rev 2024; 77:PHARMREV-AR-2024-001271. [PMID: 39406506 DOI: 10.1124/pharmrev.124.001271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 09/24/2024] [Accepted: 10/07/2024] [Indexed: 01/22/2025] Open
Abstract
Arginase catalyzes the hydrolysis of L-arginine into L-ornithine and urea. The two existing isoforms Arg1 and Arg2 show 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 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 were found 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 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 arginase, 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 The review emphasizes the need for further research to deepen our understanding of the regulation of Arg1 and Arg 2 in different cell types under consideration of their localization, species-specificity, and multiple biochemical and physiological roles. This could lead to better pharmacological strategies to target arginase activity in liver, cardiovascular, hematological, immune/infection diseases and cancer.
Collapse
Affiliation(s)
- Sophia K Heuser
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Junjie Li
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Silke Pudewell
- Department of Biochemistry and Molecular Biology II, University of Duesseldorf, Germany
| | - Anthea LoBue
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Zhixin Li
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| | - Miriam M Cortese-Krott
- Department of Cardiology, Pulmonology, and Angiology, University of Duesseldorf, Germany
| |
Collapse
|
3
|
Qiu D, Hu J, Zhang S, Cai W, Miao J, Li P, Jiang W. Fenugreek extract improves diabetes-induced endothelial dysfunction via the arginase 1 pathway. Food Funct 2024; 15:3446-3462. [PMID: 38450419 DOI: 10.1039/d3fo04283a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
Endothelial dysfunction (ED) is an initiating trigger and key factor in vascular complications, leading to disability and mortality in individuals with diabetes. The research concerning therapeutic interventions for ED has gained considerable interest. Fenugreek, a commonly used edible plant in dietary consumption, has attracted significant attention due to its management of diabetes and its associated complications. The research presented in this study examines the potential therapeutic benefits of fenugreek in treating ED and investigates the underlying mechanism associated with its effects. The analysis on fenugreek was performed using 70% ethanol extract, and its chemical composition was analyzed using ultrahigh-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). In total, we identified 49 compounds present in the fenugreek extract. These compounds encompass flavonoids, saponins, and phospholipids. Then, the models of ED in streptozotocin-induced diabetic mice and high glucose-induced isolated rat aortas were established for research. Through vascular function testing, it was observed that fenugreek extract effectively improved ED induced by diabetes or high glucose. By analyzing the protein expression of arginase 1 (Arg1), Arg activity, Arg1 immunohistochemistry, nitric oxide (NO) level, and the protein expression of endothelial nitric oxide synthase (eNOS), p38 mitogen-activated protein kinase (p38 MAPK), and p-p38 MAPK in aortas, this study revealed that the potential mechanism of fenugreek extract in anti-ED involves the downregulation of Arg1, leading to enhanced NO production. Furthermore, analysis of serum exosomes carrying Arg activity indicates that fenugreek may decrease the activity of Arg transported by serum exosomes, potentially preventing the increase in Arg levels triggered by the uptake of serum exosomes by vascular endothelial cells. In general, this investigation offers valuable observations regarding the curative impact of fenugreek extract on anti-ED in diabetes, revealing the involvement of the Arg1 pathway in its mechanism.
Collapse
Affiliation(s)
- Dingbang Qiu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
- College of Pharmacy, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Jinxin Hu
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
- College of Pharmacy, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| | - Shaoying Zhang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
| | - Wanjun Cai
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
| | - Jingwei Miao
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
| | - Pengdong Li
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
| | - Wenyue Jiang
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China.
- College of Pharmacy, Guangzhou Medical University, Guangzhou, Guangdong 511436, China
| |
Collapse
|
4
|
Gzik A, Borek B, Chrzanowski J, Jedrzejczak K, Dziegielewski M, Brzezinska J, Nowicka J, Grzybowski MM, Rejczak T, Niedzialek D, Wieczorek G, Olczak J, Golebiowski A, Zaslona Z, Blaszczyk R. Novel orally bioavailable piperidine derivatives as extracellular arginase inhibitors developed by a ring expansion. Eur J Med Chem 2024; 264:116033. [PMID: 38096651 DOI: 10.1016/j.ejmech.2023.116033] [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: 10/24/2023] [Revised: 11/24/2023] [Accepted: 12/04/2023] [Indexed: 12/30/2023]
Abstract
Arginase is a multifaced enzyme that plays an important role in health and disease being regarded as a therapeutic target for the treatment of various pathological states such as malignancies, asthma, and cardiovascular disease. The discovery of boronic acid-based arginase inhibitors in 1997 revolutionized attempts of medicinal chemistry focused on development of drugs targeting arginase. Unfortunately, these very polar compounds had limitations such as analysis and purification without chromophores, synthetically challenging space, and poor oral bioavailability. Herein, we present a novel class of boronic acid-based arginase inhibitors which are piperidine derivatives exhibiting a different pharmacological profile compared to our drug candidate in cancer immunotherapy -OATD-02 - dual ARG1/2 inhibitor with high intracellular activity. Compounds from this new series show low intracellular activity, hence they can inhibit mainly extracellular arginase, providing different therapeutic space compared to a dual intracellular ARG1/2 inhibitor. The disclosed series showed good inhibitory potential towards arginase enzyme in vitro (IC50 up to 160 nM), favorable pharmacokinetics in animal models, and encouraging preliminary in vitro and in vivo tolerability. Compounds from the new series have moderate-to-high oral bioavailability (up to 66 %) and moderate clearance in vivo. Herein we describe the development and optimization of the synthesis of the new class of boronic acid-based arginase inhibitors via a ring expansion approach starting from the inexpensive chirality source (d-hydroxyproline). This upgraded methodology facilitated a gram-scale delivery of the final compound and eliminated the need for costly and time-consuming chiral resolution.
Collapse
Affiliation(s)
- Anna Gzik
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | | | | | | | | | - Julita Nowicka
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | - Tomasz Rejczak
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | | | - Jacek Olczak
- Molecure S.A., Zwirki i Wigury 101, Warsaw, 02-089, Poland
| | | | | | | |
Collapse
|
5
|
Engin A. Endothelial Dysfunction in Obesity and Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:489-538. [PMID: 39287863 DOI: 10.1007/978-3-031-63657-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Parallel to the increasing prevalence of obesity in the world, the mortality from cardiovascular disease has also increased. Low-grade chronic inflammation in obesity disrupts vascular homeostasis, and the dysregulation of adipocyte-derived endocrine and paracrine effects contributes to endothelial dysfunction. Besides the adipose tissue inflammation, decreased nitric oxide (NO)-bioavailability, insulin resistance (IR), and oxidized low-density lipoproteins (oxLDLs) are the main factors contributing to endothelial dysfunction in obesity and the development of cardiorenal metabolic syndrome. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in the profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Higher stiffness parameter β, increased oxidative stress, upregulation of pro-inflammatory cytokines, and nicotinamide adenine dinucleotide phosphate (NADP) oxidase in PVAT turn the macrophages into pro-atherogenic phenotypes by oxLDL-induced adipocyte-derived exosome-macrophage crosstalk and contribute to the endothelial dysfunction. In clinical practice, carotid ultrasound, higher leptin levels correlate with irisin over-secretion by human visceral and subcutaneous adipose tissues, and remnant cholesterol (RC) levels predict atherosclerotic disease in obesity. As a novel therapeutic strategy for cardiovascular protection, liraglutide improves vascular dysfunction by modulating a cyclic adenosine monophosphate (cAMP)-independent protein kinase A (PKA)-AMP-activated protein kinase (AMPK) pathway in PVAT in obese individuals. Because the renin-angiotensin-aldosterone system (RAAS) activity, hyperinsulinemia, and the resultant IR play key roles in the progression of cardiovascular disease in obesity, RAAS-targeted therapies contribute to improving endothelial dysfunction. By contrast, arginase reciprocally inhibits NO formation and promotes oxidative stress. Thus, targeting arginase activity as a key mediator in endothelial dysfunction has therapeutic potential in obesity-related vascular comorbidities. Obesity-related endothelial dysfunction plays a pivotal role in the progression of type 2 diabetes (T2D). The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (thiazolidinedione), is a popular drug for treating diabetes; however, it leads to increased cardiovascular risk. Selective sodium-glucose co-transporter-2 (SGLT-2) inhibitor empagliflozin (EMPA) significantly improves endothelial dysfunction and mortality occurring through redox-dependent mechanisms. Although endothelial dysfunction and oxidative stress are alleviated by either metformin or EMPA, currently used drugs to treat obesity-related diabetes neither possess the same anti-inflammatory potential nor simultaneously target endothelial cell dysfunction and obesity equally. While therapeutic interventions with glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide or bariatric surgery reverse regenerative cell exhaustion, support vascular repair mechanisms, and improve cardiometabolic risk in individuals with T2D and obesity, the GLP-1 analog exendin-4 attenuates endothelial endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
6
|
Cross TWL, Simpson AMR, Lin CY, Hottmann NM, Bhatt AP, Pellock SJ, Nelson ER, Loman BR, Wallig MA, Vivas EI, Suchodolski J, Redinbo MR, Rey FE, Swanson KS. Gut microbiome responds to alteration in female sex hormone status and exacerbates metabolic dysfunction. Gut Microbes 2024; 16:2295429. [PMID: 38153260 PMCID: PMC10761013 DOI: 10.1080/19490976.2023.2295429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 12/12/2023] [Indexed: 12/29/2023] Open
Abstract
Women are at significantly greater risk of metabolic dysfunction after menopause, which subsequently leads to numerous chronic illnesses. The gut microbiome is associated with obesity and metabolic dysfunction, but its interaction with female sex hormone status and the resulting impact on host metabolism remains unclear. Herein, we characterized inflammatory and metabolic phenotypes as well as the gut microbiome associated with ovariectomy and high-fat diet feeding, compared to gonadal intact and low-fat diet controls. We then performed fecal microbiota transplantation (FMT) using gnotobiotic mice to identify the impact of ovariectomy-associated gut microbiome on inflammatory and metabolic outcomes. We demonstrated that ovariectomy led to greater gastrointestinal permeability and inflammation of the gut and metabolic organs, and that a high-fat diet exacerbated these phenotypes. Ovariectomy also led to alteration of the gut microbiome, including greater fecal β-glucuronidase activity. However, differential changes in the gut microbiome only occurred when fed a low-fat diet, not the high-fat diet. Gnotobiotic mice that received the gut microbiome from ovariectomized mice fed the low-fat diet had greater weight gain and hepatic gene expression related to metabolic dysfunction and inflammation than those that received intact sham control-associated microbiome. These results indicate that the gut microbiome responds to alterations in female sex hormone status and contributes to metabolic dysfunction. Identifying and developing gut microbiome-targeted modulators to regulate sex hormones may be useful therapeutically in remediating menopause-related diseases.
Collapse
Affiliation(s)
- Tzu-Wen L. Cross
- Department of Nutrition Science, Purdue University, West Lafayette, IN, USA
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Ching-Yen Lin
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Natasha M. Hottmann
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Aadra P. Bhatt
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
| | - Samuel J. Pellock
- Departments of Biochemistry & Biophysics, Microbiology & Immunology, and The Integrated Program for Biological and Genome Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Erik R. Nelson
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Cancer Center at Illinois, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Carl R. Woese Institute for Genomic Biology-Anticancer Discovery from Pets to People, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Brett R. Loman
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Matthew A. Wallig
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Eugenio I. Vivas
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jan Suchodolski
- Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, USA
| | - Matthew R. Redinbo
- Department of Chemistry, University of North Carolina, Chapel Hill, NC, USA
- Departments of Biochemistry & Biophysics, Microbiology & Immunology, and The Integrated Program for Biological and Genome Science, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Federico E. Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Cardiovascular Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Kelly S. Swanson
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
7
|
Roy R, Wilcox J, Webb AJ, O’Gallagher K. Dysfunctional and Dysregulated Nitric Oxide Synthases in Cardiovascular Disease: Mechanisms and Therapeutic Potential. Int J Mol Sci 2023; 24:15200. [PMID: 37894881 PMCID: PMC10607291 DOI: 10.3390/ijms242015200] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Nitric oxide (NO) plays an important and diverse signalling role in the cardiovascular system, contributing to the regulation of vascular tone, endothelial function, myocardial function, haemostasis, and thrombosis, amongst many other roles. NO is synthesised through the nitric oxide synthase (NOS)-dependent L-arginine-NO pathway, as well as the nitrate-nitrite-NO pathway. The three isoforms of NOS, namely neuronal (NOS1), inducible (NOS2), and endothelial (NOS3), have different localisation and functions in the human body, and are consequently thought to have differing pathophysiological roles. Furthermore, as we continue to develop a deepened understanding of the different roles of NOS isoforms in disease, the possibility of therapeutically modulating NOS activity has emerged. Indeed, impaired (or dysfunctional), as well as overactive (or dysregulated) NOS activity are attractive therapeutic targets in cardiovascular disease. This review aims to describe recent advances in elucidating the physiological role of NOS isoforms within the cardiovascular system, as well as mechanisms of dysfunctional and dysregulated NOS in cardiovascular disease. We then discuss the modulation of NO and NOS activity as a target in the development of novel cardiovascular therapeutics.
Collapse
Affiliation(s)
- Roman Roy
- Cardiovascular Department, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK;
| | - Joshua Wilcox
- Cardiovascular Department, Guy’s and St. Thomas’ NHS Foundation Trust, London SE1 7EH, UK;
| | - Andrew J. Webb
- Department of Clinical Pharmacology, British Heart Foundation Centre, School of Cardiovascular and Metabolic Medicine and Sciences, King’s College London, London SE1 7EH, UK;
| | - Kevin O’Gallagher
- Cardiovascular Department, King’s College Hospital NHS Foundation Trust, London SE5 9RS, UK;
- British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King’s College London, London SE5 9NU, UK
| |
Collapse
|
8
|
Huang YQ, Wu Z, Lin S, Chen XR. The benefits of rehabilitation exercise in improving chronic traumatic encephalopathy: recent advances and future perspectives. Mol Med 2023; 29:131. [PMID: 37740180 PMCID: PMC10517475 DOI: 10.1186/s10020-023-00728-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 09/12/2023] [Indexed: 09/24/2023] Open
Abstract
Traumatic encephalopathy syndrome (TES) is used to describe the clinical manifestations of chronic traumatic encephalopathy (CTE). However, effective treatment and prevention strategies are lacking. Increasing evidence has shown that rehabilitation training could prevent cognitive decline, enhance brain plasticity, and effectively improve neurological function in neurodegenerative diseases. Therefore, the mechanisms involved in the effects of rehabilitation exercise therapy on the prognosis of CTE are worth exploring. The aim of this article is to review the pathogenesis of CTE and provide a potential clinical intervention strategy for CTE.
Collapse
Affiliation(s)
- Yin-Qiong Huang
- Department of Endocrinology, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China
| | - Zhe Wu
- Department of Neuronal Surgery, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China.
- Group of Neuroendocrinology, Garvan Institute of Medical Research, 384 Victoria St, Sydney, Australia.
| | - Xiang-Rong Chen
- Department of Neuronal Surgery, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China.
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, No. 34 North Zhongshan Road, Quanzhou, 362000, Fujian, China.
| |
Collapse
|
9
|
Fantone S, Ermini L, Piani F, Di Simone N, Barbaro G, Giannubilo SR, Gesuita R, Tossetta G, Marzioni D. Downregulation of argininosuccinate synthase 1 (ASS1) is associated with hypoxia in placental development. Hum Cell 2023; 36:1190-1198. [PMID: 36995581 DOI: 10.1007/s13577-023-00901-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
Argininosuccinate synthase (ASS1) is involved in nitric oxide production, which has a key role in placental development improving pregnancy outcomes. Syncytiotrophoblast and extravillous trophoblast differentiations are milestones of placental development and their impairment can cause pathologies, such as preeclampsia (PE) and fetal growth restriction (FGR). Immunohistochemistry and Western blotting were used to localize and quantify ASS1 in first trimester (8.2 ± 1.8 weeks), third trimester (38.6 ± 1.1 weeks), and PE (36.3 ± 1.5 weeks) placentas. In addition, cell cultures were used to evaluate ASS1 expression under hypoxic conditions and the syncytialization process. Our data showed that ASS1 is localized in the villous cytotrophoblast of first trimester, third trimester, and PE placentas, while the villous cytotrophoblast adjacent to the extravillous trophoblast of cell columns as well as the extravillous trophoblast were negative for ASS1 in first trimester placentas. In addition, ASS1 was decreased in third trimester compared to the first trimester placentas (p = 0.003) and no differences were detected between third trimester and PE placentas. Moreover, ASS1 expression was decreased in hypoxic conditions and syncytialized cells compared to those not syncytialized. In conclusion, we suggest that the expression of ASS1 in villous cytotrophoblast is related to maintaining proliferative phenotype, while ASS1 absence may be involved in promoting the differentiation of villous cytotrophoblast in extravillous cytotrophoblast of cell columns in first trimester placentas.
Collapse
Affiliation(s)
- Sonia Fantone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Leonardo Ermini
- Department of Life Science, University of Siena, 53100, Siena, Italy
| | - Federica Piani
- Cardiovascular Internal Medicine, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40128, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, 40138, Bologna, Italy
| | - Nicoletta Di Simone
- Department of Biomedical Sciences, Humanitas University, 20072, Milan, Italy
- IRCCS Humanitas Research Hospital, 20089, Milan, Italy
| | - Greta Barbaro
- Istituto di Clinica Ostetrica e Ginecologica, Università Cattolica del Sacro Cuore, 00168, Rome, Italy
| | - Stefano Raffaele Giannubilo
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126, Ancona, Italy
| | - Rosaria Gesuita
- Centre of Epidemiology and Biostatistics, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Giovanni Tossetta
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy.
- Clinic of Obstetrics and Gynaecology, Department of Clinical Sciences, Università Politecnica delle Marche, Salesi Hospital, Azienda Ospedaliero Universitaria, 60126, Ancona, Italy.
| | - Daniela Marzioni
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| |
Collapse
|
10
|
Mahdi A, Wodaje T, Kövamees O, Tengbom J, Zhao A, Jiao T, Henricsson M, Yang J, Zhou Z, Nieminen AI, Levin M, Collado A, Brinck J, Pernow J. The red blood cell as a mediator of endothelial dysfunction in patients with familial hypercholesterolemia and dyslipidemia. J Intern Med 2023; 293:228-245. [PMID: 36324273 PMCID: PMC10092865 DOI: 10.1111/joim.13580] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Patients with familial hypercholesterolemia (FH) display high levels of low-density lipoprotein cholesterol (LDL-c), endothelial dysfunction, and increased risk of premature atherosclerosis. We have previously shown that red blood cells (RBCs) from patients with type 2 diabetes induce endothelial dysfunction through increased arginase 1 and reactive oxygen species (ROS). OBJECTIVE To test the hypothesis that RBCs from patients with FH (FH-RBCs) and elevated LDL-c induce endothelial dysfunction. METHODS AND RESULTS FH-RBCs and LDL-c >5.0 mM induced endothelial dysfunction following 18-h incubation with isolated aortic rings from healthy rats compared to FH-RBCs and LDL-c <2.5 mM or RBCs from healthy subjects (H-RBCs). Inhibition of vascular but not RBC arginase attenuated the degree of endothelial dysfunction induced by FH-RBCs and LDL-c >5.0 mM. Furthermore, arginase 1 but not arginase 2 was elevated in the vasculature of aortic segments after incubation with FH-RBCs and LDL-c >5.0 mM. A superoxide scavenger, present throughout the 18-h incubation, attenuated the degree of endothelial dysfunction induced by FH-RBCs and LDL-c >5.0 mM. ROS production was elevated in these RBCs in comparison with H-RBCs. Scavenging of vascular ROS through various antioxidants also attenuated the degree of endothelial dysfunction induced by FH-RBCs and LDL-c >5.0 mM. This was corroborated by an increase in the lipid peroxidation product 4-hydroxynonenal. Lipidomic analysis of RBC lysates did not reveal any significant changes across the groups. CONCLUSION FH-RBCs induce endothelial dysfunction dependent on LDL-c levels via arginase 1 and ROS-dependent mechanisms.
Collapse
Affiliation(s)
- Ali Mahdi
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tigist Wodaje
- Division of Cardiology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Oskar Kövamees
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Tengbom
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Allan Zhao
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Tong Jiao
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Marcus Henricsson
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Jiangning Yang
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Anni I Nieminen
- FIMM Metabolomics Unit, Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| | - Malin Levin
- Department of Molecular and Clinical Medicine, Wallenberg Laboratory, Institute of Medicine, The Sahlgrenska Academy at University of Gothenburg and Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Aida Collado
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Brinck
- Division of Endocrinology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
11
|
Li Z, Wang L, Ren Y, Huang Y, Liu W, Lv Z, Qian L, Yu Y, Xiong Y. Arginase: shedding light on the mechanisms and opportunities in cardiovascular diseases. Cell Death Dis 2022; 8:413. [PMID: 36209203 PMCID: PMC9547100 DOI: 10.1038/s41420-022-01200-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 09/17/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022]
Abstract
Arginase, a binuclear manganese metalloenzyme in the urea, catalyzes the hydrolysis of L-arginine to urea and L-ornithine. Both isoforms, arginase 1 and arginase 2 perform significant roles in the regulation of cellular functions in cardiovascular system, such as senescence, apoptosis, proliferation, inflammation, and autophagy, via a variety of mechanisms, including regulating L-arginine metabolism and activating multiple signal pathways. Furthermore, abnormal arginase activity contributes to the initiation and progression of a variety of CVDs. Therefore, targeting arginase may be a novel and promising approach for CVDs treatment. In this review, we give a comprehensive overview of the physiological and biological roles of arginase in a variety of CVDs, revealing the underlying mechanisms of arginase mediating vascular and cardiac function, as well as shedding light on the novel and promising therapeutic approaches for CVDs therapy in individuals.
Collapse
Affiliation(s)
- Zhuozhuo Li
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Liwei Wang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yuanyuan Ren
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Yaoyao Huang
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Wenxuan Liu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Ziwei Lv
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China.,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China
| | - Lu Qian
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China. .,Department of Endocrinology, Xi'an No.3 Hospital, the Affiliated Hospital of Northwest University, Northwest University, Xi'an, Shaanxi, China.
| | - Yi Yu
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China. .,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China.
| | - Yuyan Xiong
- Xi'an Key Laboratory of Cardiovascular and Cerebrovascular Diseases, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, Shaanxi, China. .,Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi'an, Shaanxi, China.
| |
Collapse
|
12
|
Heuser SK, LoBue A, Li J, Zhuge Z, Leo F, Suvorava T, Olsson A, Schneckmann R, Guimaraes Braga DD, Srivrastava T, Montero L, Schmitz OJ, Schmitt JP, Grandoch M, Weitzberg E, Lundberg JO, Pernow J, Kelm M, Carlström M, Cortese-Krott MM. Downregulation of eNOS and preserved endothelial function in endothelial-specific arginase 1-deficient mice. Nitric Oxide 2022; 125-126:69-77. [PMID: 35752264 DOI: 10.1016/j.niox.2022.06.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/08/2022] [Accepted: 06/10/2022] [Indexed: 12/20/2022]
Abstract
Arginase 1 (Arg1) is a ubiquitous enzyme belonging to the urea cycle that catalyzes the conversion of l-arginine into l-ornithine and urea. In endothelial cells (ECs), Arg1 was proposed to limit the availability of l-arginine for the endothelial nitric oxide synthase (eNOS) and thereby reduce nitric oxide (NO) production, thus promoting endothelial dysfunction and vascular disease. The role of EC Arg1 under homeostatic conditions is in vivo less understood. The aim of this study was to investigate the role of EC Arg1 on the regulation of eNOS, vascular tone, and endothelial function under normal homeostatic conditions in vivo and ex vivo. By using a tamoxifen-inducible EC-specific gene-targeting approach, we generated EC Arg1 KO mice. Efficiency and specificity of the gene targeting strategy was demonstrated by DNA recombination and loss of Arg1 expression measured after tamoxifen treatment in EC only. In EC Arg1 KO mice we found a significant decrease in Arg1 expression in heart and lung ECs and in the aorta, however, vascular enzymatic activity was preserved likely due to the presence of high levels of Arg1 in smooth muscle cells. Moreover, we found a downregulation of eNOS expression in the aorta, and a fully preserved systemic l-arginine and NO bioavailability, as demonstrated by the levels of l-arginine, l-ornithine, and l-citrulline as well as nitrite, nitrate, and nitroso-species. Lung and liver tissues from EC Arg1 KO mice showed respectively increase or decrease in nitrosyl-heme species, indicating that the lack of endothelial Arg1 affects NO bioavailability in these organs. In addition, EC Arg1 KO mice showed fully preserved acetylcholine-mediated vascular relaxation in both conductance and resistant vessels but increased phenylephrine-induced vasoconstriction. Systolic, diastolic, and mean arterial pressure and cardiac performance in EC Arg1 KO mice were not different from the wild-type littermate controls. In conclusion, under normal homeostatic conditions, lack of EC Arg1 expression is associated with a down-regulation of eNOS expression but a preserved NO bioavailability and vascular endothelial function. These results suggest that a cross-talk exists between Arg1 and eNOS to control NO production in ECs, which depends on both L-Arg availability and EC Arg1-dependent eNOS expression.
Collapse
Affiliation(s)
- Sophia K Heuser
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, 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
| | - Junjie Li
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Zhengbing Zhuge
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Francesca Leo
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tatsiana Suvorava
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Annika Olsson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Rebekka Schneckmann
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | | | - Tanu Srivrastava
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Lidia Montero
- Applied Analytical Chemistry, Faculty of Chemistry, University of Duisburg-Essen, Germany
| | - Oliver J Schmitz
- Applied Analytical Chemistry, Faculty of Chemistry, University of Duisburg-Essen, Germany
| | - Joachim P Schmitt
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Maria Grandoch
- Department of Pharmacology and Clinical Pharmacology, Medical Faculty, Heinrich-Heine-University, Germany
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - John Pernow
- Department of Cardiology, Karolinska Institute, Stockholm, Sweden
| | - Malte Kelm
- Cardiovascular Research Laboratory, Department of Cardiology Pneumology and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Miriam M Cortese-Krott
- Myocardial Infarction Research Laboratory, Department of Cardiology, Pulmonology, and Angiology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
13
|
Gajecki D, Gawryś J, Szahidewicz-Krupska E, Doroszko A. Role of Erythrocytes in Nitric Oxide Metabolism and Paracrine Regulation of Endothelial Function. Antioxidants (Basel) 2022; 11:antiox11050943. [PMID: 35624807 PMCID: PMC9137828 DOI: 10.3390/antiox11050943] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 01/27/2023] Open
Abstract
Emerging studies provide new data shedding some light on the complex and pivotal role of red blood cells (RBCs) in nitric oxide (NO) metabolism and paracrine regulation of endothelial function. NO is involved in the regulation of vasodilatation, platelet aggregation, inflammation, hypoxic adaptation, and oxidative stress. Even though tremendous knowledge about NO metabolism has been collected, the exact RBCs’ status still requires evaluation. This paper summarizes the actual knowledge regarding the role of erythrocytes as a mobile depot of amino acids necessary for NO biotransformation. Moreover, the complex regulation of RBCs’ translocases is presented with a particular focus on cationic amino acid transporters (CATs) responsible for the NO substrates and derivatives transport. The main part demonstrates the intraerythrocytic metabolism of L-arginine with its regulation by reactive oxygen species and arginase activity. Additionally, the process of nitrite and nitrate turnover was demonstrated to be another stable source of NO, with its reduction by xanthine oxidoreductase or hemoglobin. Additional function of hemoglobin in NO synthesis and its subsequent stabilization in steady intermediates is also discussed. Furthermore, RBCs regulate the vascular tone by releasing ATP, inducing smooth muscle cell relaxation, and decreasing platelet aggregation. Erythrocytes and intraerythrocytic NO metabolism are also responsible for the maintenance of normotension. Hence, RBCs became a promising new therapeutic target in restoring NO homeostasis in cardiovascular disorders.
Collapse
|
14
|
Targeting Arginine in COVID-19-Induced Immunopathology and Vasculopathy. Metabolites 2022; 12:metabo12030240. [PMID: 35323682 PMCID: PMC8953281 DOI: 10.3390/metabo12030240] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/06/2022] [Accepted: 03/09/2022] [Indexed: 01/27/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) represents a major public health crisis that has caused the death of nearly six million people worldwide. Emerging data have identified a deficiency of circulating arginine in patients with COVID-19. Arginine is a semi-essential amino acid that serves as key regulator of immune and vascular cell function. Arginine is metabolized by nitric oxide (NO) synthase to NO which plays a pivotal role in host defense and vascular health, whereas the catabolism of arginine by arginase to ornithine contributes to immune suppression and vascular disease. Notably, arginase activity is upregulated in COVID-19 patients in a disease-dependent fashion, favoring the production of ornithine and its metabolites from arginine over the synthesis of NO. This rewiring of arginine metabolism in COVID-19 promotes immune and endothelial cell dysfunction, vascular smooth muscle cell proliferation and migration, inflammation, vasoconstriction, thrombosis, and arterial thickening, fibrosis, and stiffening, which can lead to vascular occlusion, muti-organ failure, and death. Strategies that restore the plasma concentration of arginine, inhibit arginase activity, and/or enhance the bioavailability and potency of NO represent promising therapeutic approaches that may preserve immune function and prevent the development of severe vascular disease in patients with COVID-19.
Collapse
|
15
|
Mahdi A, Collado A, Tengbom J, Jiao T, Wodaje T, Johansson N, Farnebo F, Färnert A, Yang J, Lundberg JO, Zhou Z, Pernow J. Erythrocytes Induce Vascular Dysfunction in COVID-19. JACC Basic Transl Sci 2022; 7:193-204. [PMID: 35194565 PMCID: PMC8849181 DOI: 10.1016/j.jacbts.2021.12.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/03/2021] [Accepted: 12/03/2021] [Indexed: 12/20/2022]
Abstract
Patients hospitalized for COVID-19 display marked impairment in endothelial function, which is persistent following recovery from the acute infection. RBCs from patients with COVID-19 impair vascular function through mechanisms involving increased arginase 1, ROS and IFNγ, and reduced NO bioactivity. These data advance our understanding in COVID-19–associated vascular injury with a clear involvement of RBCs. Targeting these mechanisms might provide a novel therapeutic strategy to alleviate vascular injury in patients with COVID-19.
Current knowledge regarding mechanisms underlying cardiovascular complications in patients with COVID-19 is limited and urgently needed. We shed light on a previously unrecognized mechanism and unravel a key role of red blood cells, driving vascular dysfunction in patients with COVID-19 infection. We establish the presence of profound and persistent endothelial dysfunction in vivo in patients with COVID-19. Mechanistically, we show that targeting reactive oxygen species or arginase 1 improves vascular dysfunction mediated by red blood cells. These translational observations hold promise that restoring the redox balance in red blood cells might alleviate the clinical complications of COVID-19–associated vascular dysfunction.
Collapse
Key Words
- ACh, acetylcholine
- C19-RBC, red blood cell from patients with COVID-19
- COVID-19
- EDR, endothelium-dependent relaxation
- EIR, endothelium-independent relaxation
- H-RBC, red blood cell from healthy subjects
- HNE, hydroxynonenal
- IFN, interferon
- RBC, red blood cell
- RHI, reactive hyperemia index
- ROS, reactive oxygen species
- SNP, sodium nitroprusside
- TNF, tumor necrosis factor
- arginase
- endothelial dysfunction
- nitric oxide
- reactive oxygen species
- red blood cells
Collapse
Affiliation(s)
- Ali Mahdi
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Aida Collado
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Tengbom
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tong Jiao
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tigist Wodaje
- Division of Cardiology, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Niclas Johansson
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Filip Farnebo
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Stockholm Craniofacial Center, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Jiangning Yang
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Zhichao Zhou
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - John Pernow
- Division of Cardiology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
16
|
Zhou FM, Huang JJ, Hu XJ, Wang J, Zhu BQ, Ding ZS, Huang S, Fang JJ. Protective effects of flavonoids from the leaves of Carya cathayensis Sarg. against H 2O 2-induced oxidative damage and apoptosis in vitro. Exp Ther Med 2021; 22:1443. [PMID: 34721685 PMCID: PMC8549100 DOI: 10.3892/etm.2021.10878] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 09/27/2021] [Indexed: 12/19/2022] Open
Abstract
Hydrogen peroxide (H2O2) can induce apoptosis by releasing reactive oxygen species (ROS) and reactive nitrogen species, which cause mitochondrial damage. The present study aimed to investigate the protective effects of flavonoids from the leaves of Carya cathayensis Sarg. against H2O2-induced oxidative damage and apoptosis in vitro. The bioactivity of total flavonoids (TFs) and five monomeric flavonoids [cardamonin (Car), pinostrobin chalcone, wogonin, chrysin and pinocembrin] from the leaves of Carya cathayensis Sarg. (LCCS) were tested to prevent oxidative damage to rat aortic endothelial cells (RAECs) induced by H2O2. Oxidated superoxide dismutase, glutathione peroxidase, malondialdehyde, lactate dehydrogenase and ROS were analyzed to evaluate the antioxidant activity. Gene and protein expression patterns were assessed using reverse transcription-quantitative PCR and western blotting, respectively. The results indicated that TFs and Car inhibited H2O2-induced cytotoxicity and apoptosis of RAECs. Additionally, they regulated the level of oxidase and inhibited the production of ROS. Overall, the TFs extracted from LCCS could potentially be developed as effective candidate drugs to prevent oxidative stress in the future; moreover, they could also provide a direction in investigations for preventing antioxidant activity through the ROS pathway.
Collapse
Affiliation(s)
- Fang-Mei Zhou
- Technology Teaching Center of Medical Laboratory and Quarantine, School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Jing-Jing Huang
- Technology Teaching Center of Medical Laboratory and Quarantine, School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Xu-Jiao Hu
- Inspection Department, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| | - Jingwei Wang
- Department of Pathology, Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Bing-Qi Zhu
- Technology Teaching Center of Medical Laboratory and Quarantine, School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhi-Shan Ding
- Technology Teaching Center of Medical Laboratory and Quarantine, School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Shigao Huang
- Faculty of Health Sciences, University of Macau, Taipa 999078, Macau SAR, P.R. China
| | - Jing-Jing Fang
- Inspection Department, Yinzhou People's Hospital, Ningbo, Zhejiang 315040, P.R. China
| |
Collapse
|
17
|
Tengbom J, Cederström S, Verouhis D, Böhm F, Eriksson P, Folkersen L, Gabrielsen A, Jernberg T, Lundman P, Persson J, Saleh N, Settergren M, Sörensson P, Tratsiakovich Y, Tornvall P, Jung C, Pernow J. Arginase 1 is upregulated at admission in patients with ST-elevation myocardial infarction. J Intern Med 2021; 290:1061-1070. [PMID: 34237174 DOI: 10.1111/joim.13358] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The mechanisms underlying rupture of a coronary atherosclerotic plaque and development of myocardial ischemia-reperfusion injury in ST-elevation myocardial infarction (STEMI) remain unresolved. Increased arginase 1 activity leads to reduced nitric oxide (NO) production and increased formation of reactive oxygen species due to uncoupling of the NO-producing enzyme endothelial NO synthase (eNOS). This contributes to endothelial dysfunction, plaque instability and increased susceptibility to ischemia-reperfusion injury in acute myocardial infarction. OBJECTIVE The purpose of this study was to test the hypothesis that arginase gene and protein expression are upregulated in patients with STEMI. METHODS Two cohorts of patients with STEMI were included. In the first cohort (n = 51), expression of arginase and NO-synthases as well as arginase 1 protein levels were determined and compared to a healthy control group (n = 45). In a second cohort (n = 68), plasma arginase 1 levels and infarct size were determined using cardiac magnetic resonance imaging. RESULTS Expression of the gene encoding arginase 1 was significantly elevated at admission and 24-48 h after STEMI but not 3 months post STEMI, in comparison with the control group. Expression of the genes encoding arginase 2 and endothelial NO synthase (NOS3) were unaltered. Arginase 1 protein levels were elevated at admission, 24 h post STEMI and remained elevated for up to 6 months. No significant correlation between plasma arginase 1 protein levels and infarct size was observed. CONCLUSION The markedly increased gene and protein expression of arginase 1 already at admission indicates a role of arginase 1 in the development of STEMI.
Collapse
Affiliation(s)
- John Tengbom
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Sofia Cederström
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Dinos Verouhis
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Felix Böhm
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per Eriksson
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Anders Gabrielsen
- Laboratory of Immunobiology, Cardiovascular Medicine Unit, Department of Medicine, Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tomas Jernberg
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Pia Lundman
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Jonas Persson
- Department of Clinical Sciences, Karolinska Institutet, Danderyd Hospital, Stockholm, Sweden
| | - Nawzad Saleh
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Magnus Settergren
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Peder Sörensson
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Yahor Tratsiakovich
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Per Tornvall
- Department of Clinical Science and Education, Karolinska Institutet, Södersjukhuset, Stockholm, Sweden
| | - Christian Jung
- Division of Cardiology, Pulmonology, and Vascular Medicine, University Hospital Düsseldorf, Düsseldorf, Germany
| | - John Pernow
- Unit of Cardiology, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
18
|
Gilinsky MA, Polityko YK, Markel AL. Arginine Metabolism in Hypertensive Rats under Arginase Inhibition by Norvaline. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021040189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
19
|
Masi S, Rizzoni D, Taddei S, Widmer RJ, Montezano AC, Lüscher TF, Schiffrin EL, Touyz RM, Paneni F, Lerman A, Lanza GA, Virdis A. Assessment and pathophysiology of microvascular disease: recent progress and clinical implications. Eur Heart J 2021; 42:2590-2604. [PMID: 33257973 PMCID: PMC8266605 DOI: 10.1093/eurheartj/ehaa857] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/23/2020] [Accepted: 10/08/2020] [Indexed: 12/13/2022] Open
Abstract
The development of novel, non-invasive techniques and standardization of protocols to assess microvascular dysfunction have elucidated the key role of microvascular changes in the evolution of cardiovascular (CV) damage, and their capacity to predict an increased risk of adverse events. These technical advances parallel with the development of novel biological assays that enabled the ex vivo identification of pathways promoting microvascular dysfunction, providing novel potential treatment targets for preventing cerebral-CV disease. In this article, we provide an update of diagnostic testing strategies to detect and characterize microvascular dysfunction and suggestions on how to standardize and maximize the information obtained from each microvascular assay. We examine emerging data highlighting the significance of microvascular dysfunction in the development CV disease manifestations. Finally, we summarize the pathophysiology of microvascular dysfunction emphasizing the role of oxidative stress and its regulation by epigenetic mechanisms, which might represent potential targets for novel interventions beyond conventional approaches, representing a new frontier in CV disease reduction.
Collapse
Affiliation(s)
- Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.,Institute of Cardiovascular Science, University College London, London, UK
| | - Damiano Rizzoni
- Clinica Medica, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Division of Medicine, Istituto Clinico Città di Brescia, Brescia, Italy
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Robert Jay Widmer
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Augusto C Montezano
- Institute of Cardiovascular & Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Thomas F Lüscher
- Heart Division, Royal Brompton and Harefield Hospital and Imperial College, London, UK.,Center for Molecular Cardiology, University of Zürich, Zürich, Switzerland
| | - Ernesto L Schiffrin
- Department of Medicine and Lady Davis Institute, Sir Mortimer B. Davis-Jewish General Hospital, McGill University, Montreal, QC, Canada
| | - Rhian M Touyz
- Institute of Cardiovascular & Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
| | - Francesco Paneni
- Center for Molecular Cardiology, University of Zürich, Zürich, Switzerland.,Department of Cardiology, University Heart Center, University Hospital Zurich, Zürich, Switzerland.,Department of Research and Education, University Hospital Zurich, Zürich, Switzerland
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Gaetano A Lanza
- Department of Cardiovascular and Thoracic Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
20
|
Dean MJ, Ochoa JB, Sanchez-Pino MD, Zabaleta J, Garai J, Del Valle L, Wyczechowska D, Baiamonte LB, Philbrook P, Majumder R, Vander Heide RS, Dunkenberger L, Thylur RP, Nossaman B, Roberts WM, Chapple AG, Wu J, Hicks C, Collins J, Luke B, Johnson R, Koul HK, Rees CA, Morris CR, Garcia-Diaz J, Ochoa AC. Severe COVID-19 Is Characterized by an Impaired Type I Interferon Response and Elevated Levels of Arginase Producing Granulocytic Myeloid Derived Suppressor Cells. Front Immunol 2021; 12:695972. [PMID: 34341659 PMCID: PMC8324422 DOI: 10.3389/fimmu.2021.695972] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 02/06/2023] Open
Abstract
COVID-19 ranges from asymptomatic in 35% of cases to severe in 20% of patients. Differences in the type and degree of inflammation appear to determine the severity of the disease. Recent reports show an increase in circulating monocytic-myeloid-derived suppressor cells (M-MDSC) in severe COVID 19 that deplete arginine but are not associated with respiratory complications. Our data shows that differences in the type, function and transcriptome of granulocytic-MDSC (G-MDSC) may in part explain the severity COVID-19, in particular the association with pulmonary complications. Large infiltrates by Arginase 1+ G-MDSC (Arg+G-MDSC), expressing NOX-1 and NOX-2 (important for production of reactive oxygen species) were found in the lungs of patients who died from COVID-19 complications. Increased circulating Arg+G-MDSC depleted arginine, which impaired T cell receptor and endothelial cell function. Transcriptomic signatures of G-MDSC from patients with different stages of COVID-19, revealed that asymptomatic patients had increased expression of pathways and genes associated with type I interferon (IFN), while patients with severe COVID-19 had increased expression of genes associated with arginase production, and granulocyte degranulation and function. These results suggest that asymptomatic patients develop a protective type I IFN response, while patients with severe COVID-19 have an increased inflammatory response that depletes arginine, impairs T cell and endothelial cell function, and causes extensive pulmonary damage. Therefore, inhibition of arginase-1 and/or replenishment of arginine may be important in preventing/treating severe COVID-19.
Collapse
Affiliation(s)
- Matthew J. Dean
- Louisiana State University Cancer Center, New Orleans, LA, United States
| | - Juan B. Ochoa
- Department of Surgery, Ochsner Medical Center, New Orleans, LA, United States
| | - Maria Dulfary Sanchez-Pino
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Jovanny Zabaleta
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Pediatrics, LSU Health, New Orleans, LA, United States
| | - Jone Garai
- Louisiana State University Cancer Center, New Orleans, LA, United States
| | - Luis Del Valle
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Pathology LSU Health, New Orleans, LA, United States
| | | | | | - Phaethon Philbrook
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Rinku Majumder
- Department of Biochemistry, LSU Health, New Orleans, LA, United States
| | | | - Logan Dunkenberger
- Louisiana State University Cancer Center, New Orleans, LA, United States
| | | | - Bobby Nossaman
- Department of Surgery, Ochsner Medical Center, New Orleans, LA, United States
| | - W. Mark Roberts
- Department of Internal Medicine, Ochsner Medical Center, New Orleans, LA, United States
| | - Andrew G. Chapple
- Louisiana State University Cancer Center, New Orleans, LA, United States
- School of Public Health, LSU Health, New Orleans, LA, United States
| | - Jiande Wu
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Chindo Hicks
- Department of Genetics, LSU Health, New Orleans, LA, United States
| | - Jack Collins
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Brian Luke
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Randall Johnson
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, United States
| | - Hari K. Koul
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Biochemistry, LSU Health, New Orleans, LA, United States
| | - Chris A. Rees
- Division of Emergency Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Claudia R. Morris
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA, Children’s Healthcare of Atlanta, Atlanta, GA, United States
| | - Julia Garcia-Diaz
- Tissue Biorepository, Ochsner Medical Center, New Orleans, LA, United States
| | - Augusto C. Ochoa
- Louisiana State University Cancer Center, New Orleans, LA, United States
- Department of Pediatrics, LSU Health, New Orleans, LA, United States
| |
Collapse
|
21
|
Mammedova JT, Sokolov AV, Freidlin IS, Starikova EA. The Mechanisms of L-Arginine Metabolism Disorder in Endothelial Cells. BIOCHEMISTRY (MOSCOW) 2021; 86:146-155. [PMID: 33832413 DOI: 10.1134/s0006297921020036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
L-arginine is a key metabolite for nitric oxide production by endothelial cells, as well as signaling molecule of the mTOR signaling pathway. mTOR supports endothelial cells homeostasis and regulates activity of L-arginine-metabolizing enzymes, endothelial nitric oxide synthase, and arginase II. Disruption of the L-arginine metabolism in endothelial cells leads to the development of endothelial dysfunction. Conflicting results of the use of L-arginine supplement to improve endothelial function reveals a controversial role of the amino acid in the endothelial cell biology. The review is aimed at analysis of the current data on the role of L-arginine metabolism in the development of endothelial dysfunction.
Collapse
Affiliation(s)
| | - Alexey V Sokolov
- Institute of Experimental Medicine, 197376 Saint-Petersburg, Russia
| | - Irina S Freidlin
- Institute of Experimental Medicine, 197376 Saint-Petersburg, Russia
| | | |
Collapse
|
22
|
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
|
23
|
Zhu H, Wu Y, Kuang X, Liu H, Guo Z, Qian J, Wang D, Wang M, Chu H, Gong W, Zhang Z. Effect of PM 2.5 exposure on circulating fibrinogen and IL-6 levels: A systematic review and meta-analysis. CHEMOSPHERE 2021; 271:129565. [PMID: 33460893 DOI: 10.1016/j.chemosphere.2021.129565] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 12/19/2020] [Accepted: 01/04/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Ambient fine particulate matter (PM2.5) pollution poses a great threat on global health. Previous studies have reported that PM2.5 regulates circulating fibrinogen and IL-6 levels in the development of cardiovascular and respiratory disease. However, the correlation between PM2.5 exposure and both biomarkers remains inconsistent. METHODS We searched related articles through PubMed, Web of Science and ScienceDirect. Random effects model was used to obtain a pooled estimate effect of both biomarkers as PM2.5 concentration increased by every 10 μg/m3. Meta-regression analysis, sensitivity analysis and publication bias test were conducted to evaluate the heterogeneity, stability and reliability of this meta-analysis. RESULTS A total of 22 articles were included. Each 10 μg/m3 increase in PM2.5 concentration was significantly correlated with a 1.76% increase in circulating fibrinogen level (95% CI: 0.38%-3.14%, P = 0.013) and a 4.66% increase in IL-6 level (95% CI: 1.14%-8.18%, P = 0.010). Subgroup analysis revealed that high-level PM2.5 exposure had a more significant association with circulating IL-6 level (11.67%, 95% CI: 0.66%-22.69%, P = 0.038) than low-level exposure, but this association was not observed in fibrinogen (2.50%, 95% CI: -0.78%-5.77%, P = 0.135). Sensitivity analysis and publication bias test confirmed the stability of the results. CONCLUSION Circulating fibrinogen and IL-6 significantly increased with exposure to PM2.5, may serve as promising biomarkers for PM2.5-related adverse effects.
Collapse
Affiliation(s)
- Huanhuan Zhu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yanling Wu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xingya Kuang
- Department of Occupational Medicine, Yangpu Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hanting Liu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zheng Guo
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jing Qian
- Department of General Surgery, Yizheng Hospital, Nanjing Drum Tower Hospital Group, Yizheng, China
| | - Dafei Wang
- Department of Radiotherapy, Yixing Cancer Hospital, Yixing, China
| | - Meilin Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Haiyan Chu
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Weida Gong
- Department of General Surgery, Yixing People's Hospital, Yixing, China.
| | - Zhengdong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Genetic Toxicology, The Key Laboratory of Modern Toxicology of Ministry of Education, Center of Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
24
|
Dean MJ, Ochoa JB, Sanchez-Pino M, Zabaleta J, Garai J, Del Valle L, Wyczechowska D, Buckner L, Philbrook P, Majumder R, Heide RV, Dunkenberger L, Thylur R, Nossaman R, Roberts WM, Chapple A, Collins J, Luke B, Johnson R, Koul H, Rees CA, Morris CR, Garcia-Diaz J, Ochoa AC. Transcriptome and Functions of Granulocytic Myeloid-Derived Suppressor Cells Determine their Association with Disease Severity of COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021. [PMID: 33791717 DOI: 10.1101/2021.03.26.21254441] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
COVID-19 ranges from asymptomatic in 35% of cases to severe in 20% of patients. Differences in the type and degree of inflammation appear to determine the severity of the disease. Recent reports show an increase in circulating monocytic-myeloid-derived suppressor cells (M-MDSC) in severe COVID 19, that deplete arginine but are not associated with respiratory complications. Our data shows that differences in the type, function and transcriptome of Granulocytic-MDSC (G-MDSC) may in part explain the severity COVID-19, in particular the association with pulmonary complications. Large infiltrates by Arginase 1 + G-MDSC (Arg + G-MDSC), expressing NOX-1 and NOX-2 (important for production of reactive oxygen species) were found in the lungs of patients who died from COVID-19 complications. Increased circulating Arg + G-MDSC depleted arginine, which impaired T cell receptor and endothelial cell function. Transcriptomic signatures of G-MDSC from patients with different stages of COVID-19, revealed that asymptomatic patients had increased expression of pathways and genes associated with type I interferon (IFN), while patients with severe COVID-19 had increased expression of genes associated with arginase production, and granulocyte degranulation and function. These results suggest that asymptomatic patients develop a protective type I IFN response, while patients with severe COVID-19 have an increased inflammatory response that depletes arginine, impairs T cell and endothelial cell function, and causes extensive pulmonary damage. Therefore, inhibition of arginase-1 and/or replenishment of arginine may be important in preventing/treating severe COVID-19.
Collapse
|
25
|
Sabnis RW. Novel Arginase Inhibitors for Treating Cancer and Respiratory Inflammatory Diseases. ACS Med Chem Lett 2020; 11:2370-2371. [PMID: 33335657 DOI: 10.1021/acsmedchemlett.0c00593] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 12/15/2022] Open
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell & Russell LLP, 1230 Peachtree Street NE, Suite 3100, Atlanta, Georgia 30309, United States
| |
Collapse
|
26
|
Moretto J, Pudlo M, Demougeot C. Human-based evidence for the therapeutic potential of arginase inhibitors in cardiovascular diseases. Drug Discov Today 2020; 26:138-147. [PMID: 33197620 DOI: 10.1016/j.drudis.2020.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/22/2020] [Accepted: 11/05/2020] [Indexed: 01/25/2023]
Affiliation(s)
- Johnny Moretto
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France.
| | - Marc Pudlo
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| | - Céline Demougeot
- PEPITE EA4267, FHU INCREASE, Université de Bourgogne Franche-Comté, F-25030 Besançon, France
| |
Collapse
|
27
|
Erythrocytes as markers of oxidative stress related pathologies. Mech Ageing Dev 2020; 191:111333. [PMID: 32814082 DOI: 10.1016/j.mad.2020.111333] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/06/2023]
Abstract
Erythrocytes are deeply sensitive cells and important health indicators. During inflammatory response RBC, as a part of haematological system, are exposed to circulating inflammatory mediators and related oxidative stress. They present a highly specialized and organized cell membrane that interacts with inflammatory mediators and oxidative agents, leading to a variety of structural changes that promptly signal an abnormal situation. This review is aimed to provide an overview on erythrocyte involvement in physiological and pathological processes related to oxidative stress, such as aging, Down syndrome, neurodegenerative diseases, for instance Alzheimer Disease, erectile dysfunction and cardiovascular diseases. In particular this review will focus on the effects of oxidative stress on structural changes in the cell membrane and also on in the activity of erythrocyte enzymes such as membrane-bound, cytosolic glycohydrolases and RBC-eNOS. This review also underlines the potential clinical application of erythrocyte specific related parameters, which can be important tools not only for the study but also for the monitoring of several oxidative stress related diseases.
Collapse
|
28
|
Moschetta D, Di Minno MND, Porro B, Perrucci GL, Valerio V, Alfieri V, Massaiu I, Orekhov AN, Di Minno A, Songia P, Cavalca V, Myasoedova VA, Poggio P. Relationship Between Plasma Osteopontin and Arginine Pathway Metabolites in Patients With Overt Coronary Artery Disease. Front Physiol 2020; 11:982. [PMID: 32848891 PMCID: PMC7424048 DOI: 10.3389/fphys.2020.00982] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 07/20/2020] [Indexed: 11/13/2022] Open
Abstract
Introduction Osteopontin (OPN) is involved in ectopic calcification. Its circulating form is upregulated in coronary artery disease (CAD) patients. Circulating OPN levels positively correlate with oxidative stress, one of the major triggers of endothelial dysfunction. Endothelial dysfunction is, in turn, associated with reduced nitric oxide (NO) bioavailability due to the impaired arginine pathway. The aim of this study was to better understand the correlations between OPN, oxidative stress markers, and the arginine pathway metabolites. Methods and Results ELISA and mass spectrometry techniques have been used to evaluate circulating OPN and arginine pathway/oxidative stress metabolites, respectively, in twenty-five control subjects and thirty-three patients with overt atherosclerosis. OPN positively correlates with 2,3-dinor-8isoPGF2a levels (p = 0.02), ornithine (p = 0.01), ADMA (p = 0.001), SDMA (p = 0.03), and citrulline (p = 0.008) levels only in CAD patients. In addition, citrulline positively correlated with ADMA (p = 0.02) levels, possibly as result of other sources of citrulline biosynthetic pathways. Conclusion The association between OPN and impaired arginine/NO pathway could play a role in the inhibition of endothelial NO synthase (eNOS) and/or in the arginase activation in the context of CAD patients. However, further studies are needed to verify the cause-effect relationship between OPN, oxidative stress, and arginine/NO pathway dysregulation.
Collapse
Affiliation(s)
- Donato Moschetta
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milan, Italy.,Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | | | - Benedetta Porro
- Unità di Metabolomica, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Gianluca L Perrucci
- Unità di Medicina Rigenerativa e Biologia Vascolare, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Vincenza Valerio
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milan, Italy.,Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Valentina Alfieri
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Ilaria Massaiu
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Alexander N Orekhov
- Institute of General Pathology and Pathophysiology, Russian Academy of Medical Sciences, Moscow, Russia
| | - Alessandro Di Minno
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Paola Songia
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Viviana Cavalca
- Unità di Metabolomica, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Veronika A Myasoedova
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Paolo Poggio
- Unità per lo Studio delle Patologie Aortiche, Valvolari e Coronariche, Centro Cardiologico Monzino IRCCS, Milan, Italy
| |
Collapse
|
29
|
Gambardella J, Khondkar W, Morelli MB, Wang X, Santulli G, Trimarco V. Arginine and Endothelial Function. Biomedicines 2020; 8:biomedicines8080277. [PMID: 32781796 PMCID: PMC7460461 DOI: 10.3390/biomedicines8080277] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022] Open
Abstract
Arginine (L-arginine), is an amino acid involved in a number of biological processes, including the biosynthesis of proteins, host immune response, urea cycle, and nitric oxide production. In this systematic review, we focus on the functional role of arginine in the regulation of endothelial function and vascular tone. Both clinical and preclinical studies are examined, analyzing the effects of arginine supplementation in hypertension, ischemic heart disease, aging, peripheral artery disease, and diabetes mellitus.
Collapse
Affiliation(s)
- Jessica Gambardella
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine—Montefiore University Hospital, New York City, NY 10461, USA; (J.G.); (W.K.); (M.B.M.); (X.W.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York City, NY 10461, USA
- Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
- International Translational Research and Medical Education (ITME), 80100 Naples, Italy
| | - Wafiq Khondkar
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine—Montefiore University Hospital, New York City, NY 10461, USA; (J.G.); (W.K.); (M.B.M.); (X.W.)
| | - Marco Bruno Morelli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine—Montefiore University Hospital, New York City, NY 10461, USA; (J.G.); (W.K.); (M.B.M.); (X.W.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York City, NY 10461, USA
| | - Xujun Wang
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine—Montefiore University Hospital, New York City, NY 10461, USA; (J.G.); (W.K.); (M.B.M.); (X.W.)
| | - Gaetano Santulli
- Department of Medicine (Division of Cardiology), Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine—Montefiore University Hospital, New York City, NY 10461, USA; (J.G.); (W.K.); (M.B.M.); (X.W.)
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, New York City, NY 10461, USA
- Department of Advanced Biomedical Sciences, “Federico II” University, 80131 Naples, Italy
- International Translational Research and Medical Education (ITME), 80100 Naples, Italy
- Correspondence:
| | - Valentina Trimarco
- Department of Neuroscience, “Federico II” University, 80131 Naples, Italy;
| |
Collapse
|
30
|
Red Blood Cell Peroxynitrite Causes Endothelial Dysfunction in Type 2 Diabetes Mellitus via Arginase. Cells 2020; 9:cells9071712. [PMID: 32708826 PMCID: PMC7407649 DOI: 10.3390/cells9071712] [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: 06/24/2020] [Revised: 07/10/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
We recently showed that red blood cells (RBCs) from patients with type 2 diabetes mellitus (T2DM-RBCs) induce endothelial dysfunction through a mechanism involving arginase I and reactive oxygen species. Peroxynitrite is known to activate arginase in endothelial cells. Whether peroxynitrite regulates arginase activity in RBCs, and whether it is involved in the cross-talk between RBCs and the vasculature in T2DM, is unclear and elusive. The present study was designed to test the hypothesis that endothelial dysfunction induced by T2DM-RBCs is driven by peroxynitrite and upregulation of arginase. RBCs were isolated from patients with T2DM and healthy age matched controls. RBCs were co-incubated with aortae isolated from wild type rats for 18 h in the absence and presence of peroxynitrite scavenger FeTTPS. Evaluation of endothelial function in organ chambers by cumulative addition of acetylcholine as well as measurement of RBC and vessel arginase activity was performed. In another set of experiments, RBCs isolated from healthy subjects (Healthy RBCs) were incubated with the peroxynitrite donor SIN-1 with subsequent evaluation of endothelial function and arginase activity. T2DM-RBCs, but not Healthy RBCs, induced impairment in endothelial function, which was fully reversed by scavenging of RBC but not vascular peroxynitrite with FeTPPS. Arginase activity was up-regulated by the peroxynitrite donor SIN-1 in Healthy RBCs, an effect that was inhibited by FeTTPS. Healthy RBCs co-incubated with aortae in the presence of SIN-1 caused impairment of endothelial function, which was inhibited by FeTTPS or the arginase inhibitor ABH. T2DM-RBCs induced up-regulation of vascular arginase, an effect that was fully inhibited by FeTTPS. Collectively, our data indicate that RBCs impair endothelial function in T2DM via an effect that is driven by a peroxynitrite-mediated increase in arginase activity. This mechanism may be targeted in patients with T2DM for improvement in endothelial function.
Collapse
|