1
|
Gonzalez M, Clayton S, Wauson E, Christian D, Tran QK. Promotion of nitric oxide production: mechanisms, strategies, and possibilities. Front Physiol 2025; 16:1545044. [PMID: 39917079 PMCID: PMC11799299 DOI: 10.3389/fphys.2025.1545044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 01/07/2025] [Indexed: 02/09/2025] Open
Abstract
The discovery of nitric oxide (NO) and the role of endothelial cells (ECs) in its production has revolutionized medicine. NO can be produced by isoforms of NO synthases (NOS), including the neuronal (nNOS), inducible (iNOS), and endothelial isoforms (eNOS), and via the non-classical nitrate-nitrite-NO pathway. In particular, endothelium-derived NO, produced by eNOS, is essential for cardiovascular health. Endothelium-derived NO activates soluble guanylate cyclase (sGC) in vascular smooth muscle cells (VSMCs), elevating cyclic GMP (cGMP), causing vasodilation. Over the past four decades, the importance of this pathway in cardiovascular health has fueled the search for strategies to enhance NO bioavailability and/or preserve the outcomes of NO's actions. Currently approved approaches operate in three directions: 1) providing exogenous NO, 2) promoting sGC activity, and 3) preventing degradation of cGMP by inhibiting phosphodiesterase 5 activity. Despite clear benefits, these approaches face challenges such as the development of nitrate tolerance and endothelial dysfunction. This highlights the need for sustainable options that promote endogenous NO production. This review will focus on strategies to promote endogenous NO production. A detailed review of the mechanisms regulating eNOS activity will be first provided, followed by a review of strategies to promote endogenous NO production based on the levels of available preclinical and clinical evidence, and perspectives on future possibilities.
Collapse
Affiliation(s)
| | | | | | | | - Quang-Kim Tran
- Department of Physiology and Pharmacology, Des Moines University Medicine and Health Sciences, West Des Moines, IA, United States
| |
Collapse
|
2
|
You Y, Qian Z, Jiang Y, Chen L, Wu D, Liu L, Zhang F, Ning X, Zhang Y, Xiao J. Insights into the pathogenesis of gestational and hepatic diseases: the impact of ferroptosis. Front Cell Dev Biol 2024; 12:1482838. [PMID: 39600338 PMCID: PMC11588751 DOI: 10.3389/fcell.2024.1482838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
Ferroptosis, a distinct form of non-apoptotic cell death characterized by iron dependency and lipid peroxidation, is increasingly linked to various pathological conditions in pregnancy and liver diseases. It plays a critical role throughout pregnancy, influencing processes such as embryogenesis, implantation, and the maintenance of gestation. A growing body of evidence indicates that disruptions in these processes can precipitate pregnancy-related disorders, including pre-eclampsia (PE), gestational diabetes mellitus (GDM), and intrahepatic cholestasis of pregnancy (ICP). Notably, while ICP is primarily associated with elevated maternal serum bile acid levels, its precise etiology remains elusive. Oxidative stress induced by bile acid accumulation is believed to be a significant factor in ICP pathogenesis. Similarly, the liver's susceptibility to oxidative damage underscores the importance of lipid metabolism dysregulation and impaired iron homeostasis in the progression of liver diseases such as alcoholic liver disease (ALD), non-alcoholic fatty liver disease (NAFLD), cholestatic liver injury, autoimmune hepatitis (AIH), acute liver injury, viral hepatitis, liver fibrosis, and hepatocellular carcinoma (HCC). This review discusses the shared signaling mechanisms of ferroptosis in gestational and hepatic diseases, and explores recent advances in understanding the mechanisms of ferroptosis and its potential role in the pathogenesis of gestational and hepatic disorders, with the aim of identifying viable therapeutic targets.
Collapse
Affiliation(s)
- Yilan You
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Zhiwen Qian
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Ying Jiang
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Lingyan Chen
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Danping Wu
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Lu Liu
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Feng Zhang
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Xin Ning
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Yan Zhang
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| | - Jianping Xiao
- Departments of Obstetrics and Gynecology, Wuxi Maternal and Child Healthcare Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
- Departments of Obstetrics and Gynecology, Wuxi Maternity and Child Healthcare Hospital, Women’s Hospital of Jiangnan University, Jiangnan University, Wuxi, China
| |
Collapse
|
3
|
Pun CK, Huang HC, Chang CC, Chuang CL, Hsu SJ, Hou MC, Lee FY. Fructooligosaccharides reverses hepatic vascular dysfunction and dysbiosis in rats with liver cirrhosis and portal hypertension. Eur J Clin Invest 2024; 54:e14287. [PMID: 39017981 DOI: 10.1111/eci.14287] [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: 04/15/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
BACKGROUND Portal hypertension leads to lethal complications in liver cirrhosis. Oxidative stress induced hepatic vascular dysfunction, which exaggerated vasoconstriction and increases hepatic vascular resistance (HVR). Gut dysbiosis further exacerbates portal hypertension. Fructooligosaccharides are prebiotics with potent antioxidant effect. This study aimed to evaluate the roles of fructooligosaccharides in portal hypertension-related vascular dysregulation and gut microbiome. METHODS Sprague-Dawley rats received bile duct ligation to induce cirrhosis or sham operation as controls. The rats then randomly received fructooligosaccharides or vehicle for 4 weeks. Experiments were performed on the 29th day after operations. RESULTS Fructooligosaccharides did not affect portal pressure. Interestingly, fructooligosaccharides significantly attenuated HVR (p = .03). Malondialdehyde, an oxidative stress marker, reduced significantly in the liver in fructooligosaccharides-treated group. In addition, superoxide dismutase and trolox equivalent antioxidant capacity increased in the treatment group. On the other hand, vasodilatation-related protein expressions, GTPCH and phospho-eNOS, enhanced significantly. Fructooligosaccharides had no adverse vasodilatation effects on splanchnic vascular system or porto-systemic collateral systems. Locomotor function was not affected by fructooligosaccharides. Faecal microbiota analysis showed that Negativicutes, Selenomonadales and Lactobacillus salivarius reduced in the fructooligosaccharides-treated group. CONCLUSION In conclusion, fructooligosaccharides attenuate hepatic vascular dysfunction in cirrhotic rats via at least partly, ameliorate of dysbiosis and oxidative stress.
Collapse
Affiliation(s)
- Chon Kit Pun
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Therapeutic and Research Center of Liver Cirrhosis and Portal Hypertension, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Hui-Chun Huang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Therapeutic and Research Center of Liver Cirrhosis and Portal Hypertension, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ching-Chih Chang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Holistic and Multidisciplinary Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Chiao-Lin Chuang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of General Medicine, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shao-Jung Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Therapeutic and Research Center of Liver Cirrhosis and Portal Hypertension, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ming-Chih Hou
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Therapeutic and Research Center of Liver Cirrhosis and Portal Hypertension, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Fa-Yauh Lee
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Therapeutic and Research Center of Liver Cirrhosis and Portal Hypertension, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
4
|
Duță C, Muscurel C, Dogaru CB, Stoian I. Ferroptosis-A Shared Mechanism for Parkinson's Disease and Type 2 Diabetes. Int J Mol Sci 2024; 25:8838. [PMID: 39201524 PMCID: PMC11354749 DOI: 10.3390/ijms25168838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/31/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Type 2 diabetes (T2D) and Parkinson's disease (PD) are the two most frequent age-related chronic diseases. There are many similarities between the two diseases: both are chronic diseases; both are the result of a decrease in a specific substance-insulin in T2D and dopamine in PD; and both are caused by the destruction of specific cells-beta pancreatic cells in T2D and dopaminergic neurons in PD. Recent epidemiological and experimental studies have found that there are common underlying mechanisms in the pathophysiology of T2D and PD: chronic inflammation, mitochondrial dysfunction, impaired protein handling and ferroptosis. Epidemiological research has indicated that there is a higher risk of PD in individuals with T2D. Moreover, clinical studies have observed that the symptoms of Parkinson's disease worsen significantly after the onset of T2D. This article provides an up-to-date review on the intricate interplay between oxidative stress, reactive oxygen species (ROS) and ferroptosis in PD and T2D. By understanding the shared molecular pathways and how they can be modulated, we can develop more effective therapies, or we can repurpose existing drugs to improve patient outcomes in both disorders.
Collapse
|
5
|
Xia W, Zhang M, Liu C, Wang S, Xu A, Xia Z, Pang L, Cai Y. Exploring the therapeutic potential of tetrahydrobiopterin for heart failure with preserved ejection fraction: A path forward. Life Sci 2024; 345:122594. [PMID: 38537900 DOI: 10.1016/j.lfs.2024.122594] [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: 01/17/2024] [Revised: 03/10/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
A large number of patients are affected by classical heart failure (HF) symptomatology with preserved ejection fraction (HFpEF) and multiorgan syndrome. Due to high morbidity and mortality rate, hospitalization and mortality remain serious socioeconomic problems, while the lack of effective pharmacological or device treatment means that HFpEF presents a major unmet medical need. Evidence from clinical and basic studies demonstrates that systemic inflammation, increased oxidative stress, and impaired mitochondrial function are the common pathological mechanisms in HFpEF. Tetrahydrobiopterin (BH4), beyond being an endogenous co-factor for catalyzing the conversion of some essential biomolecules, has the capacity to prevent systemic inflammation, enhance antioxidant resistance, and modulate mitochondrial energy production. Therefore, BH4 has emerged in the last decade as a promising agent to prevent or reverse the progression of disorders such as cardiovascular disease. In this review, we cover the clinical progress and limitations of using downstream targets of nitric oxide (NO) through NO donors, soluble guanylate cyclase activators, phosphodiesterase inhibitors, and sodium-glucose co-transporter 2 inhibitors in treating cardiovascular diseases, including HFpEF. We discuss the use of BH4 in association with HFpEF, providing new evidence for its potential use as a pharmacological option for treating HFpEF.
Collapse
Affiliation(s)
- Weiyi Xia
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China
| | - Miao Zhang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Guangdong, China
| | - Chang Liu
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China
| | - Sheng Wang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; Department of Medicine, The University of Hong Kong, Hong Kong SAR, China; Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Guangdong, China
| | - Lei Pang
- Department of Anesthesiology, The First Hospital of Jilin University, Jilin, China.
| | - Yin Cai
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Center for Chinese Medicine Innovation, The Hong Kong Polytechnic University, Hong Kong SAR, China; Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong SAR, China.
| |
Collapse
|
6
|
Lu L, Jang S, Zhu J, Qin Q, Sun L, Sun J. Nur77 mitigates endothelial dysfunction through activation of both nitric oxide production and anti-oxidant pathways. Redox Biol 2024; 70:103056. [PMID: 38290383 PMCID: PMC10844745 DOI: 10.1016/j.redox.2024.103056] [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: 10/17/2023] [Revised: 01/05/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Nur77 belongs to the member of orphan nuclear receptor 4A family that plays critical roles in maintaining vascular homeostasis. This study aims to determine whether Nur77 plays a role in attenuating vascular dysfunction, and if so, to determine the molecular mechanisms involved. METHODS Both Nur77 knockout (Nur77 KO) and Nur77 endothelial specific transgenic mice (Nur77-Tg) were employed to examine the functional significance of Nur77 in vascular endothelium in vivo. Endothelium-dependent vasodilatation to acetylcholine (Ach) and reactive oxygen species (ROS) production was determined under inflammatory and high glucose conditions. Expression of genes was determined by real-time PCR and western blot analysis. RESULTS In response to tumor necrosis factor alpha (TNF-α) treatment and diabetes, the endothelium-dependent vasodilatation to Ach was significantly impaired in aorta from Nur77 KO as compared with those from the wild-type (WT) mice. Endothelial specific overexpression of Nur77 markedly prevented both TNF-α- and high glucose-induced endothelial dysfunction. Compared with WT mice, after TNF-α and high glucose treatment, ROS production in aorta was significantly increased in Nur77 KO mice, but it was inhibited in Nur77-Tg mice, as determined by dihydroethidium (DHE) staining. Furthermore, we demonstrated that Nur77 overexpression substantially increased the expression of several key enzymes involved in nitric oxide (NO) production and ROS scavenging, including endothelial nitric oxide synthase (eNOS), guanosine triphosphate cyclohydrolase 1 (GCH-1), glutathione peroxidase-1 (GPx-1), and superoxide dismutases (SODs). Mechanistically, we found that Nur77 increased GCH1 mRNA stability by inhibiting the expression of microRNA-133a, while Nur77 upregulated SOD1 expression through directly binding to the human SOD1 promoter in vascular endothelial cells. CONCLUSION Our results suggest that Nur77 plays an essential role in attenuating endothelial dysfunction through activating NO production and anti-oxidant pathways in vascular endothelium. Targeted activation of Nur77 may provide a novel therapeutic approach for the treatment of cardiovascular diseases associated with endothelial dysfunction.
Collapse
Affiliation(s)
- Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, PR China
| | - Soohwa Jang
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Jiaqi Zhu
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Qing Qin
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Lijun Sun
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Jianxin Sun
- Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA, 19107, USA.
| |
Collapse
|
7
|
Cinicola BL, Palumbo IM, Pannunzio A, Carnevale R, Bartimoccia S, Cammisotto V, Capponi M, Brindisi G, Salvatori F, Barillà F, Martino F, D'Amico A, Poscia R, Spalice A, Zicari AM, Violi F, Loffredo L. Low Grade Endotoxemia and Oxidative Stress in Offspring of Patients with Early Myocardial Infarction. Antioxidants (Basel) 2023; 12:antiox12040958. [PMID: 37107333 PMCID: PMC10135978 DOI: 10.3390/antiox12040958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/14/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Background and aims: Offspring of patients with early myocardial infarction are at higher cardiovascular risk, but the underlying physio-pathological mechanism is unclear. NADPH oxidase-type 2 (NOX-2) plays a pivotal role as mediator of oxidative stress and could be involved in activating platelets in these patients. Furthermore, altered intestinal permeability and serum lipopolysaccharide (LPS) could be a trigger to promote NOX-2 activation and platelet aggregation. This study aims to evaluate the behavior of low grade endotoxemia, oxidative stress and platelet activation in offspring of patients with early myocardial infarction. Methods: We enrolled, in a cross-sectional study, 46 offspring of patients with early myocardial infarction and 86 healthy subjects (HS). LPS levels and gut permeability (assessed by zonulin), oxidative stress (assessed by serum NOX-2-derived peptide (sNOX2-dp) release, hydrogen peroxide (H2O2) production and isoprostanes), serum nitric oxide (NO) bioavailability and platelet activation (by serum thromboxane B2 (TXB2) and soluble P-Selectin (sP-Selectin)) were analyzed. Results: Compared to HS, offspring of patients with early myocardial infarction had higher values of LPS, zonulin, serum isoprostanes, sNOX2-dp H2O2, TXB2, p-selectin and lower NO bioavailability. Logistic regression analysis showed that the variables associated with offspring of patients with early myocardial infarction were LPS, TXB2 and isoprostanes. The multiple linear regression analysis confirmed that serum NOX-2, isoprostanes, p-selectin and H2O2 levels were significantly associated to LPS. Furthermore, serum LPS, isoprostanes and TXB2 levels were significantly associated with sNOX-2-dp. Conclusions: Offspring of patients with early myocardial infarction have a low grade endotoxemia that could generate oxidative stress and platelet activation increasing their cardiovascular risk. Future studies are needed to understand the role of dysbiosis in this population.
Collapse
Affiliation(s)
- Bianca Laura Cinicola
- Department of Maternal Infantile and Urological Sciences, Division of Pediatric Allergology and Immunology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 391, 00161 Rome, Italy
| | - Ilaria Maria Palumbo
- Department of Clinical, Internistic, Anaesthetic and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Arianna Pannunzio
- Department of Clinical, Internistic, Anaesthetic and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Corso della Repubblica, 79, 04100 Latina, Italy
- IRCCS Neuromed, Località Camerele, 86077 Pozzilli, Italy
| | - Simona Bartimoccia
- Department of Clinical, Internistic, Anaesthetic and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Vittoria Cammisotto
- Department of Clinical, Internistic, Anaesthetic and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Martina Capponi
- Department of Maternal Infantile and Urological Sciences, Division of Pediatric Allergology and Immunology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Giulia Brindisi
- Department of Maternal Infantile and Urological Sciences, Division of Pediatric Allergology and Immunology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Francesca Salvatori
- Department of Maternal Infantile and Urological Sciences, Division of Pediatric Allergology and Immunology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Francesco Barillà
- Unit of Cardiology, University Hospital "Tor Vergata", 00133 Rome, Italy
| | - Francesco Martino
- Department of Pediatrics and Pediatric Neuropsychiatry, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
| | - Alessandra D'Amico
- Department of Movement, Human and Health Sciences, University of Rome "Foro Italico", Piazza Lauro De Bosis, 15, 00135 Rome, Italy
| | - Roberto Poscia
- Unita di Ricerca Clinica e Clinical Competence-Direzione Generale, AOU Policlinico Umberto I, 00161 Rome, Italy
| | - Alberto Spalice
- Department of Maternal Infantile and Urological Sciences, Division of Pediatric Allergology and Immunology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Anna Maria Zicari
- Department of Maternal Infantile and Urological Sciences, Division of Pediatric Allergology and Immunology, Sapienza University of Rome, Viale Regina Elena 324, 00161 Rome, Italy
| | - Francesco Violi
- Department of Pediatrics and Pediatric Neuropsychiatry, Sapienza University of Rome, Piazzale Aldo Moro, 5, 00185 Rome, Italy
- Mediterranea Cardiocentro-Napoli, 80122 Naples, Italy
| | - Lorenzo Loffredo
- Department of Clinical, Internistic, Anaesthetic and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| |
Collapse
|
8
|
Janaszak-Jasiecka A, Płoska A, Wierońska JM, Dobrucki LW, Kalinowski L. Endothelial dysfunction due to eNOS uncoupling: molecular mechanisms as potential therapeutic targets. Cell Mol Biol Lett 2023; 28:21. [PMID: 36890458 PMCID: PMC9996905 DOI: 10.1186/s11658-023-00423-2] [Citation(s) in RCA: 54] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/19/2023] [Indexed: 03/10/2023] Open
Abstract
Nitric oxide (NO) is one of the most important molecules released by endothelial cells, and its antiatherogenic properties support cardiovascular homeostasis. Diminished NO bioavailability is a common hallmark of endothelial dysfunction underlying the pathogenesis of the cardiovascular disease. Vascular NO is synthesized by endothelial nitric oxide synthase (eNOS) from the substrate L-arginine (L-Arg), with tetrahydrobiopterin (BH4) as an essential cofactor. Cardiovascular risk factors such as diabetes, dyslipidemia, hypertension, aging, or smoking increase vascular oxidative stress that strongly affects eNOS activity and leads to eNOS uncoupling. Uncoupled eNOS produces superoxide anion (O2-) instead of NO, thus becoming a source of harmful free radicals exacerbating the oxidative stress further. eNOS uncoupling is thought to be one of the major underlying causes of endothelial dysfunction observed in the pathogenesis of vascular diseases. Here, we discuss the main mechanisms of eNOS uncoupling, including oxidative depletion of the critical eNOS cofactor BH4, deficiency of eNOS substrate L-Arg, or accumulation of its analog asymmetrical dimethylarginine (ADMA), and eNOS S-glutathionylation. Moreover, potential therapeutic approaches that prevent eNOS uncoupling by improving cofactor availability, restoration of L-Arg/ADMA ratio, or modulation of eNOS S-glutathionylation are briefly outlined.
Collapse
Affiliation(s)
- Anna Janaszak-Jasiecka
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Agata Płoska
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland
| | - Joanna M Wierońska
- Department of Neurobiology, Polish Academy of Sciences, Maj Institute of Pharmacology, 12 Smętna Street, 31-343, Kraków, Poland
| | - Lawrence W Dobrucki
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Beckman Institute for Advanced Science and Technology, 405 N Mathews Ave, MC-251, Urbana, IL, 61801, USA.,Department of Biomedical and Translational Sciences, Carle-Illinois College of Medicine, Urbana, IL, USA
| | - Leszek Kalinowski
- Department of Medical Laboratory Diagnostics - Fahrenheit Biobank BBMRI.Pl, Medical University of Gdansk, 7 Debinki Street, 80-211, Gdansk, Poland. .,BioTechMed Centre, Department of Mechanics of Materials and Structures, Gdansk University of Technology, 11/12 Gabriela Narutowicza Street, 80-233, Gdansk, Poland.
| |
Collapse
|
9
|
Suppression of GCH1 Sensitizes Ovarian Cancer and Breast Cancer to PARP Inhibitor. JOURNAL OF ONCOLOGY 2023; 2023:1453739. [PMID: 36793373 PMCID: PMC9925261 DOI: 10.1155/2023/1453739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/26/2022] [Accepted: 11/24/2022] [Indexed: 02/08/2023]
Abstract
Background Breast and ovarian cancers are common malignancies among women, contributing to a significant disease burden, and are characterized by a high level of genomic instability, owing to the failure of homologous recombination repair (HRR). Pharmacological inhibition of poly(ADP-ribose) polymerase (PARP) could elicit the synthetic lethal effect of tumor cells in patients with homologous recombination deficiency, ultimately achieving a favorable clinical benefit. However, primary and acquired resistance remain the greatest hurdle, limiting the efficacy of PARP inhibitors; thus, strategies conferring or augmenting tumor cell sensitivity to PARP inhibitors are urgently required. Methods Our RNA-seq data of niraparib-treated and -untreated tumor cells were analyzed by R language. Gene Set Enrichment Analysis (GSEA) was applied to assess the biological functions of GTP cyclohydrolase 1 (GCH1). Quantitative real-time PCR, Western blotting, and immunofluorescence were applied to confirm the upregulation of GCH1 upon niraparib treatment at transcriptional and translational levels. Immunohistochemistry of patient-derived xenograft (PDX)-derived tissue sections further validated that niraparib increased GCH1 expression. Tumor cell apoptosis was detected by flow cytometry, while the superiority of the combination strategy was confirmed in the PDX model. Results The expression of GCH1 was aberrantly enriched in breast and ovarian cancers and increased after niraparib treatment via JAK-STAT signaling. GCH1 was also demonstrated to be associated with the HRR pathway. Subsequently, the enhancement of the tumor-killing effect of PARP inhibitors induced by GCH1 suppression using siRNA and GCH1 inhibitor was validated by flow cytometry in vitro. Finally, using the PDX model, we further demonstrated that GCH1 inhibitors markedly potentiated PARP inhibitors' antitumor efficacy in vivo. Conclusion Our results illustrated that PARP inhibitors promote GCH1 expression via the JAK-STAT pathway. We also elucidated the potential relationship between GCH1 and the homologous recombination repair pathway and proposed a combination regimen of GCH1 suppression with PARP inhibitors in breast and ovarian cancers.
Collapse
|
10
|
Sonmez MI, Shahzadi A, Kose C, Sonmez H, Ozyazgan S, Akkan AG. Effect of sulfasalazine on endothelium-dependent vascular response by the activation of Nrf2 signalling pathway. Front Pharmacol 2022; 13:979300. [PMID: 36353481 PMCID: PMC9639785 DOI: 10.3389/fphar.2022.979300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/29/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Diabetes mellitus leads to endothelial dysfunction and accumulation of oxygen radicals. Sulfasalazine-induced Nrf2 activation reduces oxidative stress in vessels. Thus, in the present study, we investigated the effects of sulfasalazine on endothelial dysfunction induced by high glucose. We also ascribed the underlying mechanism involved in glucose-induced endothelial dysfunction. Methods: For this experiment we used 80 Wistar Albino rats thoracic aorta to calculate the dose response curve of noradrenaline and acetylcholine. Vessels were incubated in normal and high glucose for 2 h. To investigate glucose and sulfasalazine effects the vessels of the high glucose group were pre-treated with sulfasalazine (300 mM), JNK inhibitor (SP600125), and ERK inhibitor (U0126) for 30 min. The dose response curve was calculated through organ bath. The eNOS, TAS, TOS, and HO-1 levels were estimated by commercially available ELISA kits. Results: In the high glucose group, the Emax for contraction was significantly higher (p < 0.001), and Emax for relaxation was lower than that of control. These functional changes were parallel with the low levels of eNOS (p < 0.05). High glucose vessel treated with sulfasalazine showed low Emax value for contraction (p < 0.001) however, the Emax for relaxation was significantly high (p < 0.001) when compared to high glucose group. In the JNK group, Emax for contraction and relaxation was inhibited (p < 0.001) compared to sulfasalazine treated vessels. HO—1 enzyme levels were significantly low (p < 0.01) with sulfasalazine but higher with ERK inhibitor (p < 0.05). Conclusion: High glucose induced endothelial dysfunction and sulfasalazine reduced damage in high glucose vessels by activating eNOS, antioxidant effect through HO-1 enzymes and particularly inducing Nrf2 via the ERK and JNK pathways.
Collapse
Affiliation(s)
- Muhammed Ikbal Sonmez
- Institute of Experimental Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
- DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
- *Correspondence: Muhammed Ikbal Sonmez,
| | - Andleeb Shahzadi
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Cagla Kose
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
- Department of Medical Pharmacology, Medical Faculty, Halic University, Istanbul, Turkey
| | - Haktan Sonmez
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Sibel Ozyazgan
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Ahmet Gokhan Akkan
- Department of Medical Pharmacology, Cerrahpasa Medical Faculty, Istanbul University-Cerrahpasa, Istanbul, Turkey
- Department of Medical Pharmacology, Medical Faculty, Bezmialem Vakif University Hospital, Istanbul, Turkey
| |
Collapse
|
11
|
Zhang Z, Dalan R, Hu Z, Wang JW, Chew NW, Poh KK, Tan RS, Soong TW, Dai Y, Ye L, Chen X. Reactive Oxygen Species Scavenging Nanomedicine for the Treatment of Ischemic Heart Disease. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2202169. [PMID: 35470476 DOI: 10.1002/adma.202202169] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/08/2022] [Indexed: 06/14/2023]
Abstract
Ischemic heart disease (IHD) is the leading cause of disability and mortality worldwide. Reactive oxygen species (ROS) have been shown to play key roles in the progression of diabetes, hypertension, and hypercholesterolemia, which are independent risk factors that lead to atherosclerosis and the development of IHD. Engineered biomaterial-based nanomedicines are under extensive investigation and exploration, serving as smart and multifunctional nanocarriers for synergistic therapeutic effect. Capitalizing on cell/molecule-targeting drug delivery, nanomedicines present enhanced specificity and safety with favorable pharmacokinetics and pharmacodynamics. Herein, the roles of ROS in both IHD and its risk factors are discussed, highlighting cardiovascular medications that have antioxidant properties, and summarizing the advantages, properties, and recent achievements of nanomedicines that have ROS scavenging capacity for the treatment of diabetes, hypertension, hypercholesterolemia, atherosclerosis, ischemia/reperfusion, and myocardial infarction. Finally, the current challenges of nanomedicines for ROS-scavenging treatment of IHD and possible future directions are discussed from a clinical perspective.
Collapse
Affiliation(s)
- Zhan Zhang
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 408433, Singapore
| | - Zhenyu Hu
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jiong-Wei Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Nicholas Ws Chew
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Kian-Keong Poh
- Department of Cardiology, National University Heart Centre, National University Hospital, Singapore, 119074, Singapore
| | - Ru-San Tan
- Department of Cardiology, National Heart Centre Singapore, Singapore, 119609, Singapore
| | - Tuck Wah Soong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Yunlu Dai
- Cancer Centre and Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
- MoE Frontiers Science Center for Precision Oncology, University of Macao, Taipa, Macau SAR, 999078, China
| | - Lei Ye
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology and Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Chemical and Biomolecular Engineering and Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, 117597, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| |
Collapse
|
12
|
Measurement of Tetrahydrobiopterin in Animal Tissue Samples by HPLC with Electrochemical Detection-Protocol Optimization and Pitfalls. Antioxidants (Basel) 2022; 11:antiox11061182. [PMID: 35740082 PMCID: PMC9228106 DOI: 10.3390/antiox11061182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/10/2022] [Accepted: 06/14/2022] [Indexed: 11/17/2022] Open
Abstract
Tetrahydrobiopterin (BH4) is an essential cofactor of all nitric oxide synthase isoforms, thus determination of BH4 levels can provide important mechanistic insight into diseases. We established a protocol for high-performance liquid chromatography/electrochemical detection (HPLC/ECD)-based determination of BH4 in tissue samples. We first determined the optimal storage and work-up conditions for authentic BH4 and its oxidation product dihydrobiopterin (BH2) under various conditions (pH, temperature, presence of antioxidants, metal chelators, and storage time). We then applied optimized protocols for detection of BH4 in tissues of septic (induced by lipopolysaccharide [LPS]) rats. BH4 standards in HCl are stabilized by addition of 1,4-dithioerythritol (DTE) and diethylenetriaminepentaacetic acid (DTPA), while HCl was sufficient for BH2 standard stabilization. Overnight storage of BH4 standard solutions at room temperature in HCl without antioxidants caused complete loss of BH4 and the formation of BH2. We further optimized the protocol to separate ascorbate and the BH4 tissue sample and found a significant increase in BH4 in the heart and kidney as well as higher BH4 levels by trend in the brain of septic rats compared to control rats. These findings correspond to reports on augmented nitric oxide and BH4 levels in both animals and patients with septic shock.
Collapse
|
13
|
Sumi-Ichinose C, Suganuma Y, Kano T, Ikemoto K, Ihira N, Ichinose H, Kondo K. Priapism caused by partial deficiency of tetrahydrobiopterin through hypofunction of the sympathetic neurons in sepiapterin reductase gene-disrupted mice. J Inherit Metab Dis 2022; 45:621-634. [PMID: 35192730 DOI: 10.1002/jimd.12489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 02/03/2022] [Accepted: 02/16/2022] [Indexed: 11/11/2022]
Abstract
6R-L-erythro-5,6,7,8-tetrahydrobiopterin (BH4) is an essential cofactor for aromatic L-amino acid hydroxylases, including tyrosine hydroxylase (TH), alkylglycerol monooxygenase, and three types of nitric oxide (NO) synthases (NOS). Sepiapterin reductase (SPR) catalyzes the third step of BH4 biosynthesis. SPR gene-disrupted (Spr-/- ) mice exhibit a dystonic posture, low body weight, hyperphenylalaninemia, and unstable hypertension with endothelial dysfunction. In this study, we found that Spr-/- mice suffered from a high incidence of severe priapism. Their erections persisted for months. The biopterin, BH4, and norepinephrine contents, and TH protein levels in the penile tissue of Spr-/- mice without and with priapism were significantly reduced compared to those of Spr+/+ mice. In contrast, their neural NOS (nNOS) protein levels were increased, and the cyclic guanosine monophosphate (cGMP) levels were remarkably elevated in the penises of Spr-/- mice with priapism. The symptoms were relieved by repeated administration of BH4. The biopterin, BH4, and norepinephrine contents were increased in penile homogenates from BH4-supplemented Spr-/- mice, and the TH protein levels tended to increase, and their nitrite plus nitrate levels were significantly lower than those of vehicle-treated Spr-/- mice and were approximately the same as vehicle- and BH4-supplemented Spr+/+ mice. Thus, we deduced that the priapism of Spr-/- mice is primarily caused by hypofunction of the sympathetic neurons due to cofactor depletion and the loss of TH protein and, further, dysregulation of the NO/cGMP signaling pathway, which would be caused by disinhibition of nNOS-containing neurons and/or abnormal catabolism of cyclic nucleotides is suggested.
Collapse
Affiliation(s)
- Chiho Sumi-Ichinose
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Yui Suganuma
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Taiki Kano
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Kazuhisa Ikemoto
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Noriko Ihira
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| | - Hiroshi Ichinose
- School of Life Science and Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Kazunao Kondo
- Department of Pharmacology, School of Medicine, Fujita Health University, Toyoake, Japan
| |
Collapse
|
14
|
Reily-Bell M, Bahn A, Katare R. Reactive Oxygen Species-Mediated Diabetic Heart Disease: Mechanisms and Therapies. Antioxid Redox Signal 2022; 36:608-630. [PMID: 34011169 DOI: 10.1089/ars.2021.0098] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Significance: Diabetic heart disease (DHD) is the primary cause of mortality in people with diabetes. A significant contributor to the development of DHD is the disruption of redox balance due to reactive oxygen species (ROS) overproduction resulting from sustained high glucose levels. Therapies specifically focusing on the suppression of ROS will hugely benefit patients with DHD. Recent Advances: In addition to the gold standard pharmacological therapies, the recent development of gene therapy provides an exciting avenue for developing new therapeutics to treat ROS-mediated DHD. In particular, microRNAs (miRNAs) are gaining interest due to their crucial role in several physiological and pathological processes, including DHD. Critical Issues: miRNAs have many targets and differential function depending on the environment. Therefore, a proper understanding of the function of miRNAs in specific cell types and cell states is required for the successful application of this technology. In the present review, we first provide an overview of the role of ROS in contributing to DHD and the currently available treatments. We then discuss the newer gene therapies with a specific focus on the role of miRNAs as the causative factors and therapeutic targets to combat ROS-mediated DHD. Future Directions: The future of miRNA therapeutics in tackling ROS-mediated DHD is dependent on a complete understanding of how miRNAs behave in different cells and environments. Future research should also aim to develop conditional miRNA therapeutic platforms capable of switching on and off in response to disruptions in the redox state. Antioxid. Redox Signal. 36, 608-630.
Collapse
Affiliation(s)
- Matthew Reily-Bell
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - Andrew Bahn
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| | - Rajesh Katare
- Department of Physiology-HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
15
|
Sen A, Singh A, Roy A, Mohanty S, Naik N, Kalaivani M, Ramakrishnan L. Role of endothelial colony forming cells (ECFCs) Tetrahydrobiopterin (BH4) in determining ECFCs functionality in coronary artery disease (CAD) patients. Sci Rep 2022; 12:3076. [PMID: 35197509 PMCID: PMC8866483 DOI: 10.1038/s41598-022-06758-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Accepted: 01/31/2022] [Indexed: 01/05/2023] Open
Abstract
Nitric oxide (NO.) is critical for functionality of endothelial colony forming cells (ECFCs). Dimerization of endothelial nitric oxide synthase (eNOS) is must to produce NO. and tetrahydrobiopterin (BH4) plays a crucial role in stabilizing this state. We investigated BH4 level in ECFCs and its effect on ECFCs functionality in CAD patients. Intracellular biopterin levels and ECFCs functionality in terms of cell viability, adhesion, proliferation, in vitro wound healing and angiogenesis were assessed. Guanosine Triphosphate Cyclohydrolase-1 (GTPCH-1) expression was studied in ECFCs. Serum total reactive oxygen/nitrogen species was measured and effect of nitrosative stress on ECFC's biopterins level and functionality were evaluated by treating with 3-morpholino sydnonimine (SIN-1). BH4 level was significantly lower in ECFCs from CAD patients. Cell proliferation, wound closure reflecting cellular migration as well as in vitro angiogenesis were impaired in ECFCs from CAD patients. Wound healing capacity and angiogenesis were positively correlated with ECFC's BH4. A negative effect of nitrosative stress on biopterins level and cell functionality was observed in SIN-1 treated ECFCs. ECFCs from CAD exhibited impaired functionality and lower BH4 level. Association of BH4 with wound healing capacity and angiogenesis suggest its role in maintaining ECFC's functionality. Oxidative stress may be a determinant of intracellular biopterin levels.
Collapse
Affiliation(s)
- Atanu Sen
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archna Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Ambuj Roy
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Sujata Mohanty
- Centre of Excellence for Stem Cell Research, All India Institute of Medical Sciences, New Delhi, India
| | - Nitish Naik
- Department of Cardiology, All India Institute of Medical Sciences, New Delhi, India
| | - Mani Kalaivani
- Department of Biostatistics, All India Institute of Medical Sciences, New Delhi, India
| | - Lakshmy Ramakrishnan
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
16
|
El-Mahdy MA, Ewees MG, Eid MS, Mahgoup EM, Khaleel SA, Zweier JL. Electronic Cigarette Exposure Causes Vascular Endothelial Dysfunction Due to NADPH Oxidase Activation and eNOS Uncoupling. Am J Physiol Heart Circ Physiol 2022; 322:H549-H567. [PMID: 35089811 PMCID: PMC8917923 DOI: 10.1152/ajpheart.00460.2021] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
We recently reported a mouse model of chronic electronic cigarette (e-cig) exposure-induced cardiovascular pathology, where long-term exposure to e-cig vape (ECV) induces cardiac abnormalities, impairment of endothelial function, and systemic hypertension. Here, we delineate the underlying mechanisms of ECV-induced vascular endothelial dysfunction (VED), a central trigger of cardiovascular disease. C57/BL6 male mice were exposed to ECV generated from e-cig liquid containing 0, 6, or 24 mg/ml nicotine for 16 and 60 weeks. Time-dependent elevation in blood pressure and systemic vascular resistance were observed, along with an impairment of acetylcholine-induced aortic relaxation in ECV-exposed mice, compared to air-exposed control. Decreased intravascular nitric oxide (NO) levels and increased superoxide generation with elevated 3-nitrotyrosine levels in the aorta of ECV-exposed mice were observed, indicating that ECV-induced superoxide reacts with NO to generate cytotoxic peroxynitrite. Exposure increased NADPH oxidase expression, supporting its role in ECV-induced superoxide generation. Downregulation of endothelial nitric oxide synthase (eNOS) expression and Akt-dependent eNOS phosphorylation occurred in the aorta of ECV-exposed mice, indicating that exposure inhibited de novo NO synthesis. Following ECV exposure, the critical NOS cofactor tetrahydrobiopterin was decreased, with a concomitant loss of its salvage enzyme, dihydrofolate reductase. NADPH oxidase and NOS inhibitors abrogated ECV-induced superoxide generation in the aorta of ECV exposed mice. Together, our data demonstrate that ECV exposure activates NADPH oxidase and uncouples eNOS, causing a vicious cycle of superoxide generation and vascular oxidant stress that triggers VED and hypertension with predisposition to other cardiovascular disease.
Collapse
Affiliation(s)
- Mohamed A El-Mahdy
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Mohamed G Ewees
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Mahmoud S Eid
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Elsayed M Mahgoup
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Sahar A Khaleel
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States.,Department of Pharmacology and Toxicology, College of Pharmacy, Al-Azhar University, Cairo, Egypt
| | - Jay L Zweier
- Center for Environmental and Smoking Induced Disease and the Department of Internal Medicine, Division of Cardiovascular Medicine, Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
17
|
Khairallah A, Ross CJ, Tastan Bishop Ö. GTP Cyclohydrolase I as a Potential Drug Target: New Insights into Its Allosteric Modulation via Normal Mode Analysis. J Chem Inf Model 2021; 61:4701-4719. [PMID: 34450011 DOI: 10.1021/acs.jcim.1c00898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Guanosine triphosphate (GTP) cyclohydrolase I (GCH1) catalyzes the conversion of GTP into dihydroneopterin triphosphate (DHNP). DHNP is the first intermediate of the folate de novo biosynthesis pathway in prokaryotic and lower eukaryotic microorganisms and the tetrahydrobiopterin (BH4) biosynthesis pathway in higher eukaryotes. The de novo folate biosynthesis provides essential cofactors for DNA replication, cell division, and synthesis of key amino acids in rapidly replicating pathogen cells, such as Plasmodium falciparum (P. falciparum), a causative agent of malaria. In eukaryotes, the product of the BH4 biosynthesis pathway is essential for the production of nitric oxide and several neurotransmitter precursors. An increased copy number of the malaria parasite P. falciparum GCH1 gene has been reported to influence antimalarial antifolate drug resistance evolution, whereas mutations in the human GCH1 are associated with neuropathic and inflammatory pain disorders. Thus, GCH1 stands as an important and attractive drug target for developing therapeutics. The GCH1 intrinsic dynamics that modulate its activity remains unclear, and key sites that exert allosteric effects across the structure are yet to be elucidated. This study employed the anisotropic network model to analyze the intrinsic motions of the GCH1 structure alone and in complex with its regulatory partner protein. We showed that the GCH1 tunnel-gating mechanism is regulated by a global shear motion and an outward expansion of the central five-helix bundle. We further identified hotspot residues within sites of structural significance for the GCH1 intrinsic allosteric modulation. The obtained results can provide a solid starting point to design novel antineuropathic treatments for humans and novel antimalarial drugs against the malaria parasite P. falciparum GCH1 enzyme.
Collapse
Affiliation(s)
- Afrah Khairallah
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown 6140, South Africa
| | - Caroline J Ross
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown 6140, South Africa
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Grahamstown 6140, South Africa
| |
Collapse
|
18
|
Veitch CR, Power AS, Erickson JR. CaMKII Inhibition is a Novel Therapeutic Strategy to Prevent Diabetic Cardiomyopathy. Front Pharmacol 2021; 12:695401. [PMID: 34381362 PMCID: PMC8350113 DOI: 10.3389/fphar.2021.695401] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/14/2021] [Indexed: 11/24/2022] Open
Abstract
Increasing prevalence of diabetes mellitus worldwide has pushed the complex disease state to the foreground of biomedical research, especially concerning its multifaceted impacts on the cardiovascular system. Current therapies for diabetic cardiomyopathy have had a positive impact, but with diabetic patients still suffering from a significantly greater burden of cardiac pathology compared to the general population, the need for novel therapeutic approaches is great. A new therapeutic target, calcium/calmodulin-dependent kinase II (CaMKII), has emerged as a potential treatment option for preventing cardiac dysfunction in the setting of diabetes. Within the last 10 years, new evidence has emerged describing the pathophysiological consequences of CaMKII activation in the diabetic heart, the mechanisms that underlie persistent CaMKII activation, and the protective effects of CaMKII inhibition to prevent diabetic cardiomyopathy. This review will examine recent evidence tying cardiac dysfunction in diabetes to CaMKII activation. It will then discuss the current understanding of the mechanisms by which CaMKII activity is enhanced during diabetes. Finally, it will examine the benefits of CaMKII inhibition to treat diabetic cardiomyopathy, including contractile dysfunction, heart failure with preserved ejection fraction, and arrhythmogenesis. We intend this review to serve as a critical examination of CaMKII inhibition as a therapeutic strategy, including potential drawbacks of this approach.
Collapse
Affiliation(s)
- Christopher R Veitch
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Amelia S Power
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| | - Jeffrey R Erickson
- Department of Physiology and HeartOtago, University of Otago, Dunedin, New Zealand
| |
Collapse
|
19
|
Contreras-Duarte S, Claudette C, Farias M, Leiva A. High total cholesterol and triglycerides levels increase arginases metabolism, impairing nitric oxide signaling and worsening fetoplacental endothelial dysfunction in gestational diabetes mellitus pregnancies. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166216. [PMID: 34314821 DOI: 10.1016/j.bbadis.2021.166216] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/18/2021] [Accepted: 07/07/2021] [Indexed: 12/23/2022]
Abstract
During human pregnancy, maternal physiological dyslipidemia (MPD) supports fetal development. However, some women develop maternal supraphysiological dyslipidemia (MSPD: increased total cholesterol (TC) and triglycerides (TG) levels). MSPD is present in normal and pregnancies with gestational diabetes mellitus (GDM). Both pathologies associate with fetoplacental endothelial dysfunction, producing alterations in nitric oxide (NO)-L-arginine/arginase metabolism. Nevertheless, the effect of MSPD on GDM, and how this synergy alters fetoplacental endothelial function is unknown, which is the aim of this study. 123 women at term of pregnancy were classified as MPD (n=40), MSPD (n=35), GDM with normal lipids (GDM- MPD, n=23) and with increased lipids (GDM-MSPD, n=25). TC ≥291 mg/dL and TG ≥275 mg/dL were considered as MSPD. Endothelial NO synthase (eNOS), human cationic amino acid transporter 1 (hCat1), and arginase II protein abundance and activity, were assayed in umbilical vein endothelial cells. In MSPD and MSPD-GDM, TC and TG increased respect to MPD and MPD-GDM. eNOS activity was reduced in MSPD and MSPD-GDM, but increased in MPD-GDM compared with MPD. No changes were observed in eNOS protein. However, decreased tetrahydrobiopterin levels were observed in all groups compared with MPD. Increased hCat1 protein and L-arginine transport were observed in both GDM groups compared with MPD. However, the transport was higher in GDM-MSPD compared to GDM-MPD. Higher Arginase II protein and activity were observed in MSPD-GDM compared with MPD. Thus, MSPD in GDM pregnancies alters fetal endothelial function associated with NO metabolism.
Collapse
Affiliation(s)
- S Contreras-Duarte
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago 7510156, Chile.
| | - C Claudette
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago 7510156, Chile
| | - M Farias
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastian, Santiago 7510156, Chile
| | - A Leiva
- Department of Obstetrics, School of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile.
| |
Collapse
|
20
|
Himmelreich N, Blau N, Thöny B. Molecular and metabolic bases of tetrahydrobiopterin (BH 4) deficiencies. Mol Genet Metab 2021; 133:123-136. [PMID: 33903016 DOI: 10.1016/j.ymgme.2021.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 01/01/2023]
Abstract
Tetrahydrobiopterin (BH4) deficiency is caused by genetic variants in the three genes involved in de novo cofactor biosynthesis, GTP cyclohydrolase I (GTPCH/GCH1), 6-pyruvoyl-tetrahydropterin synthase (PTPS/PTS), sepiapterin reductase (SR/SPR), and the two genes involved in cofactor recycling, carbinolamine-4α-dehydratase (PCD/PCBD1) and dihydropteridine reductase (DHPR/QDPR). Dysfunction in BH4 metabolism leads to reduced cofactor levels and may result in systemic hyperphenylalaninemia and/or neurological sequelae due to secondary deficiency in monoamine neurotransmitters in the central nervous system. More than 1100 patients with BH4 deficiency and 800 different allelic variants distributed throughout the individual genes are tabulated in database of pediatric neurotransmitter disorders PNDdb. Here we provide an update on the molecular-genetic analysis and structural considerations of these variants, including the clinical courses of the genotypes. From a total of 324 alleles, 11 are associated with the autosomal recessive form of GTPCH deficiency presenting with hyperphenylalaninemia (HPA) and neurotransmitter deficiency, 295 GCH1 variant alleles are detected in the dominant form of L-dopa-responsive dystonia (DRD or Segawa disease) while phenotypes of 18 alleles remained undefined. Autosomal recessive variants observed in the PTS (199 variants), PCBD1 (32 variants), and QDPR (141 variants) genes lead to HPA concomitant with central monoamine neurotransmitter deficiency, while SPR deficiency (104 variants) presents without hyperphenylalaninemia. The clinical impact of reported variants is essential for genetic counseling and important for development of precision medicine.
Collapse
Affiliation(s)
- Nastassja Himmelreich
- Center for Child and Adolescent Medicine, Dietmar-Hopp Metabolic Center, Division 1, Heidelberg, Germany
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital Zürich, Zürich, Switzerland.
| | - Beat Thöny
- Division of Metabolism and Children's Research Centre, University Children's Hospital Zürich, Zürich, Switzerland.
| |
Collapse
|
21
|
Deng J. Research progress on the molecular mechanism of coronary microvascular endothelial cell dysfunction. IJC HEART & VASCULATURE 2021; 34:100777. [PMID: 33912653 PMCID: PMC8065195 DOI: 10.1016/j.ijcha.2021.100777] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/27/2021] [Accepted: 03/30/2021] [Indexed: 12/19/2022]
Abstract
Coronary microvascular disease is a high-risk factor for many cardiovascular events. However, due to its high concealment and many etiologies, the current understanding of its pathophysiological mechanism is very limited, which greatly limits its clinical diagnosis and treatment. In the process of the occurrence and development of coronary microvascular disease, the damage of coronary microvascular endothelial cell (CMEC) is the core link. CMEC's stress, metabolism, inflammation and other dysfunctions have a causal relationship with coronary microvascular disease, and are also the main features of coronary microvascular disease in the early stage. This article mainly reviews the molecular mechanisms of CMEC damage.
Collapse
Affiliation(s)
- Jianying Deng
- Department of Cardiovascular Surgery, Chongqing Kanghua Zhonglian Cardiovascular Hospital, Chong Qing, China
| |
Collapse
|
22
|
Carnicer R, Duglan D, Ziberna K, Recalde A, Reilly S, Simon JN, Mafrici S, Arya R, Rosello-Lleti E, Chuaiphichai S, Tyler D, Lygate CA, Channon KM, Casadei B. BH4 Increases nNOS Activity and Preserves Left Ventricular Function in Diabetes. Circ Res 2021; 128:585-601. [PMID: 33494625 PMCID: PMC7612785 DOI: 10.1161/circresaha.120.316656] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 01/25/2021] [Indexed: 12/20/2022]
Abstract
RATIONALE In diabetic patients, heart failure with predominant left ventricular (LV) diastolic dysfunction is a common complication for which there is no effective treatment. Oxidation of the NOS (nitric oxide synthase) cofactor tetrahydrobiopterin (BH4) and dysfunctional NOS activity have been implicated in the pathogenesis of the diabetic vascular and cardiomyopathic phenotype. OBJECTIVE Using mice models and human myocardial samples, we evaluated whether and by which mechanism increasing myocardial BH4 availability prevented or reversed LV dysfunction induced by diabetes. METHODS AND RESULTS In contrast to the vascular endothelium, BH4 levels, superoxide production, and NOS activity (by liquid chromatography) did not differ in the LV myocardium of diabetic mice or in atrial tissue from diabetic patients. Nevertheless, the impairment in both cardiomyocyte relaxation and [Ca2+]i (intracellular calcium) decay and in vivo LV function (echocardiography and tissue Doppler) that developed in wild-type mice 12 weeks post-diabetes induction (streptozotocin, 42-45 mg/kg) was prevented in mGCH1-Tg (mice with elevated myocardial BH4 content secondary to trangenic overexpression of GTP-cyclohydrolase 1) and reversed in wild-type mice receiving oral BH4 supplementation from the 12th to the 18th week after diabetes induction. The protective effect of BH4 was abolished by CRISPR/Cas9-mediated knockout of nNOS (the neuronal NOS isoform) in mGCH1-Tg. In HEK (human embryonic kidney) cells, S-nitrosoglutathione led to a PKG (protein kinase G)-dependent increase in plasmalemmal density of the insulin-independent glucose transporter GLUT-1 (glucose transporter-1). In cardiomyocytes, mGCH1 overexpression induced a NO/sGC (soluble guanylate cyclase)/PKG-dependent increase in glucose uptake via GLUT-1, which was instrumental in preserving mitochondrial creatine kinase activity, oxygen consumption rate, LV energetics (by 31phosphorous magnetic resonance spectroscopy), and myocardial function. CONCLUSIONS We uncovered a novel mechanism whereby myocardial BH4 prevents and reverses LV diastolic and systolic dysfunction associated with diabetes via an nNOS-mediated increase in insulin-independent myocardial glucose uptake and utilization. These findings highlight the potential of GCH1/BH4-based therapeutics in human diabetic cardiomyopathy. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
| | - Drew Duglan
- Cardiovascular Medicine, University of Oxford
| | | | | | | | | | | | - Ritu Arya
- Cardiovascular Medicine, University of Oxford
| | | | | | - Damian Tyler
- Physiology, Anatomy and Genetics, University of Oxford
| | | | | | | |
Collapse
|
23
|
Panday S, Kar S, Kavdia M. How does ascorbate improve endothelial dysfunction? - A computational analysis. Free Radic Biol Med 2021; 165:111-126. [PMID: 33497797 DOI: 10.1016/j.freeradbiomed.2021.01.031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 12/23/2020] [Accepted: 01/14/2021] [Indexed: 01/02/2023]
Abstract
Low levels of ascorbate (Asc) are observed in cardiovascular and neurovascular diseases. Asc has therapeutic potential for the treatment of endothelial dysfunction, which is characterized by a reduction in nitric oxide (NO) bioavailability and increased oxidative stress in the vasculature. However, the potential mechanisms remain poorly understood for the Asc mitigation of endothelial dysfunction. In this study, we developed an endothelial cell based computational model integrating endothelial cell nitric oxide synthase (eNOS) biochemical pathway with downstream reactions and interactions of oxidative stress, tetrahydrobiopterin (BH4) synthesis and biopterin ratio ([BH4]/[TBP]), Asc and glutathione (GSH). We quantitatively analyzed three Asc mediated mechanisms that are reported to improve/maintain endothelial cell function. The mechanisms include the reduction of •BH3 to BH4, direct scavenging of superoxide (O2•-) and peroxynitrite (ONOO-) and increasing eNOS activity. The model predicted that Asc at 0.1-100 μM concentrations improved endothelial cell NO production, total biopterin and biopterin ratio in a dose dependent manner and the extent of cellular oxidative stress. Asc increased BH4 availability and restored eNOS coupling under oxidative stress conditions. Asc at concentrations of 1-10 mM reduced O2•- and ONOO- levels and could act as an antioxidant. We predicted that glutathione peroxidase and peroxiredoxin in combination with GSH and Asc can restore eNOS coupling and NO production under oxidative stress conditions. Asc supplementation may be used as an effective therapeutic strategy when BH4 levels are depleted. This study provides detailed understanding of the mechanism responsible and the optimal cellular Asc levels for improvement in endothelial dysfunction.
Collapse
Affiliation(s)
- Sheetal Panday
- Department of Biomedical Engineering, Wayne State University, Detroit, 48202, MI, USA
| | - Saptarshi Kar
- College of Engineering and Technology, American University of the Middle East, Kuwait
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, 48202, MI, USA.
| |
Collapse
|
24
|
Yan L, Zhang JQZ, Dai XY, Li JF, Gao FY, Zhang XF, Tian YJ, Shi WJ, Zhu JB, Chen JK. Endothelium-Specific GTP Cyclohydrolase I Overexpression Restores Endothelial Function in Aged Mice. J Vasc Res 2021; 58:1-5. [PMID: 33556943 DOI: 10.1159/000513464] [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: 06/18/2020] [Accepted: 11/26/2020] [Indexed: 11/19/2022] Open
Abstract
This study tested the hypothesis that endothelium-specific GTP cyclohydrolase I (GTPCH I) overexpression (Tg-GCH) restores age-associated endothelial dysfunction in vivo. Aortic GTPCH I expression and serum nitric oxide (NO) release were measured in young and aged mice. Aortic rings from young and aged wild-type (WT) mice and aged Tg-GCH mice were suspended for isometric tension recording. A hind limb ischemia model was used to measure blood flow recovery. Aged mice showed reduced GTPCH I expression in the aorta and decreased NO levels in serum. Compared with aged WT mice, Tg-GCH significantly elevated NO levels in serum in aged Tg-GCH mice, restored the impaired aortic relaxation in response to acetylcholine, and significantly elevated aortic constriction in response to L-NAME. Importantly, aged Tg-GCH mice displayed a significant increase in blood flow recovery compared with aged WT mice. GTPCH I reduction contributes to aging-associated endothelial dysfunction, which can be retarded by Tg-GCH.
Collapse
Affiliation(s)
- Lang Yan
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Ji-Qian-Zhu Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xiao-Yu Dai
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Jin-Feng Li
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Fang-Yuan Gao
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Xiao-Fang Zhang
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Yi-Jun Tian
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Wen-Jing Shi
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Jiang-Bo Zhu
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China
| | - Ji-Kuai Chen
- Department of Health Toxicology, Faculty of Naval Medicine, Second Military Medical University, Shanghai, China,
| |
Collapse
|
25
|
Sailer S, Keller MA, Werner ER, Watschinger K. The Emerging Physiological Role of AGMO 10 Years after Its Gene Identification. Life (Basel) 2021; 11:life11020088. [PMID: 33530536 PMCID: PMC7911779 DOI: 10.3390/life11020088] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/21/2021] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
Abstract
The gene encoding alkylglycerol monooxygenase (AGMO) was assigned 10 years ago. So far, AGMO is the only known enzyme capable of catalysing the breakdown of alkylglycerols and lyso-alkylglycerophospholipids. With the knowledge of the genetic information, it was possible to relate a potential contribution for mutations in the AGMO locus to human diseases by genome-wide association studies. A possible role for AGMO was implicated by genetic analyses in a variety of human pathologies such as type 2 diabetes, neurodevelopmental disorders, cancer, and immune defence. Deficient catabolism of stored lipids carrying an alkyl bond by an absence of AGMO was shown to impact on the overall lipid composition also outside the ether lipid pool. This review focuses on the current evidence of AGMO in human diseases and summarises experimental evidence for its role in immunity, energy homeostasis, and development in humans and several model organisms. With the progress in lipidomics platform and genetic identification of enzymes involved in ether lipid metabolism such as AGMO, it is now possible to study the consequence of gene ablation on the global lipid pool and further on certain signalling cascades in a variety of model organisms in more detail.
Collapse
Affiliation(s)
- Sabrina Sailer
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
| | - Markus A. Keller
- Institute of Human Genetics, Medical University of Innsbruck, 6020 Innsbruck, Austria;
| | - Ernst R. Werner
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
| | - Katrin Watschinger
- Biocenter, Institute of Biological Chemistry, Medical University of Innsbruck, 6020 Innsbruck, Austria; (S.S.); (E.R.W.)
- Correspondence: ; Tel.: +43-512-9003-70344
| |
Collapse
|
26
|
Abstract
Generation of nitric oxide (NO) by the nitric oxide synthase (NOS) enzymes plays multiple signalling roles in every organ system, with crucial roles in the cardiovascular system, mediated by endothelial nitric oxide synthase (eNOS, encoded by NOS3) and neuronal nitric oxide synthase (nNOS, encoded by NOS1) in regulation of blood pressure, flow, oxygen delivery and cardiac function. Loss of normal NO-mediated functions in cardiovascular disease state is associated with changes in nitroso-redox signalling that are not dependent solely upon altered NO generation, but increased generation of reactive oxygen species (ROS). The NOS enzymes can also generate ROS, in a catalytic mode whereby the generation of NO from L-arginine is 'uncoupled' from the reduction of molecular oxygen. NOS uncoupling is determined by several factors, including the availability and oxidation state of the required NOS cofactor, tetrahydrobiopterin (BH4). The duality of NOS functions as enzymes that generate both NO and ROS under different regulatory states has emerged as an important pathophysiologic mechanism, and is a potential therapeutic target, via agents that can maintain or restore NOS coupling, for example via effects on BH4 availability.
Collapse
Affiliation(s)
- Keith M Channon
- BHF Field Marshal Earl Alexander Professor of Cardiovascular Medicine, University of Oxford and Oxford University Hospitals, Oxford, UK.
| |
Collapse
|
27
|
Alexander Y, Osto E, Schmidt-Trucksäss A, Shechter M, Trifunovic D, Duncker DJ, Aboyans V, Bäck M, Badimon L, Cosentino F, De Carlo M, Dorobantu M, Harrison DG, Guzik TJ, Hoefer I, Morris PD, Norata GD, Suades R, Taddei S, Vilahur G, Waltenberger J, Weber C, Wilkinson F, Bochaton-Piallat ML, Evans PC. Endothelial function in cardiovascular medicine: a consensus paper of the European Society of Cardiology Working Groups on Atherosclerosis and Vascular Biology, Aorta and Peripheral Vascular Diseases, Coronary Pathophysiology and Microcirculation, and Thrombosis. Cardiovasc Res 2021; 117:29-42. [PMID: 32282914 PMCID: PMC7797212 DOI: 10.1093/cvr/cvaa085] [Citation(s) in RCA: 185] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/08/2020] [Accepted: 04/07/2020] [Indexed: 12/14/2022] Open
Abstract
Endothelial cells (ECs) are sentinels of cardiovascular health. Their function is reduced by the presence of cardiovascular risk factors, and is regained once pathological stimuli are removed. In this European Society for Cardiology Position Paper, we describe endothelial dysfunction as a spectrum of phenotypic states and advocate further studies to determine the role of EC subtypes in cardiovascular disease. We conclude that there is no single ideal method for measurement of endothelial function. Techniques to measure coronary epicardial and micro-vascular function are well established but they are invasive, time-consuming, and expensive. Flow-mediated dilatation (FMD) of the brachial arteries provides a non-invasive alternative but is technically challenging and requires extensive training and standardization. We, therefore, propose that a consensus methodology for FMD is universally adopted to minimize technical variation between studies, and that reference FMD values are established for different populations of healthy individuals and patient groups. Newer techniques to measure endothelial function that are relatively easy to perform, such as finger plethysmography and the retinal flicker test, have the potential for increased clinical use provided a consensus is achieved on the measurement protocol used. We recommend further clinical studies to establish reference values for these techniques and to assess their ability to improve cardiovascular risk stratification. We advocate future studies to determine whether integration of endothelial function measurements with patient-specific epigenetic data and other biomarkers can enhance the stratification of patients for differential diagnosis, disease progression, and responses to therapy.
Collapse
Affiliation(s)
- Yvonne Alexander
- Centre for Bioscience, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | - Elena Osto
- Institute of Clinical Chemistry, University and University Hospital Zurich, University Heart Center, Zurich, Switzerland
- Laboratory of Translational Nutrition Biology, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Arno Schmidt-Trucksäss
- Division of Sports and Exercise Medicine, Department of Sport, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland
| | - Michael Shechter
- Leviev Heart Center, Chaim Sheba Medical Center, Tel Hashomer, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Danijela Trifunovic
- Cardiology Department, Clinical Centre of Serbia, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Victor Aboyans
- Department of Cardiology, Dupuytren University Hospital, Inserm U-1094, Limoges University, Limoges, France
| | - Magnus Bäck
- Department of Cardiology, Center for Molecular Medicine, Karolinska University Hospital, Solna, Stockholm, Sweden
- INSERM U1116, Université de Lorraine, Centre Hospitalier Régional Universitaire de Nancy, Vandoeuvre les Nancy, France
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Francesco Cosentino
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Marco De Carlo
- Catheterization Laboratory, Azienda Ospedaliero-Universitaria Pisana, Pisa, Italy
| | - Maria Dorobantu
- ‘CarolDavila’ University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Department of Medicine, Jagiellonian University Collegium Medicum, Cracow, Poland
| | - Imo Hoefer
- Laboratory of Clinical Chemistry and Hematology, University Medical Centre Utrecht, The Netherlands
| | - Paul D Morris
- Department of Infection, Immunity and Cardiovascular Disease, Bateson Centre & INSIGNEO Institute, University of Sheffield, Sheffield S10 2RX, UK
- Insigneo Institute for In Silico Medicine, Sheffield, UK
| | - Giuseppe D Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Rosa Suades
- Unit of Cardiology, Karolinska Institute and Karolinska University Hospital, Solna, Stockholm, Sweden
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, IR-Hospital de la Santa Creu i Sant Pau, CiberCV, Autonomous University of Barcelona, Barcelona, Spain
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, Medical Faculty, University of Münster, Münster, Germany
- SRH Central Hospital Suhl, Suhl, Germany
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillian-Universität (LMU) München, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Fiona Wilkinson
- Centre for Bioscience, Faculty of Science & Engineering, Manchester Metropolitan University, Manchester, UK
| | | | - Paul C Evans
- Department of Infection, Immunity and Cardiovascular Disease, Bateson Centre & INSIGNEO Institute, University of Sheffield, Sheffield S10 2RX, UK
- Insigneo Institute for In Silico Medicine, Sheffield, UK
| |
Collapse
|
28
|
Zhu ML, Gao ZT, Lu JX, Wang Y, Wang G, Zhu TT, Li P, Liu C, Wang SX, Yang L. Amorphous nano-selenium quantum dots prevent pulmonary arterial hypertension through recoupling endothelial nitric oxide synthase. Aging (Albany NY) 2020; 13:3368-3385. [PMID: 33323558 PMCID: PMC7906187 DOI: 10.18632/aging.202215] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/29/2020] [Indexed: 04/28/2023]
Abstract
AIMS We have previously reported that nano-selenium quantum dots (SeQDs) prevented endothelial dysfunction in atherosclerosis. This study is to investigate whether amorphous SeQDs (A-SeQDs) increase endogenous tetrahydrobiopterin biosynthesis to alleviate pulmonary arterial hypertension. RESULTS Both A-SeQDs and C-SeQDs were stable under physiological conditions, while the size of A-SeQDs was smaller than C-SeQDs by high resolution-transmission electron microscopy scanning. In monocrotaline-injected mice, oral administration of A-SeQDs was more effective to decrease pulmonary arterial pressure, compared to C-SeQDs and organic selenium. Further, A-SeQDs increased both nitric oxide productions and intracellular BH4 levels, upregulated dihydrofolate reductase activity in lungs, and improved pulmonary arterial remodeling. Gene deletion of dihydrofolate reductase abolished these effects produced by A-SeQDs in mice. Finally, the blood levels of tetrahydrobiopterin and selenium were decreased in patients with pulmonary arterial hypertension. CONCLUSION A-SeQDs increase intracellular tetrahydrobiopterin to prevent pulmonary arterial hypertension through recoupling endothelial nitric oxide synthase. METHODS Two polymorphs of SeQDs and A-SeQDs, and a crystalline form of SeQDs (C-SeQDs) were prepared through self-redox decomposition of selenosulfate precursor. Mice were injected with monocrotaline to induce pulmonary arterial hypertension in vivo. Pulmonary arterial pressure was measured.
Collapse
Affiliation(s)
- Mo-Li Zhu
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Zhi-Tao Gao
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jun-Xiu Lu
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yang Wang
- Department of Pharmacy, The 3rd Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Ge Wang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China
| | - Tian-Tian Zhu
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Peng Li
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Shuang-Xi Wang
- Henan International Joint Laboratory of Cardiovascular Remodeling and Drug Intervention, School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, Hubei, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lin Yang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China
| |
Collapse
|
29
|
Seppan P, Muhammed I, Mohammad ZIK, Sathyanathan SB. Pathobiology of ischiocavernosus and bulbospongiosus muscles in long-term diabetic male rats and its implication on erectile dysfunction. Aging Male 2020; 23:979-990. [PMID: 31368398 DOI: 10.1080/13685538.2019.1647160] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
OBJECTIVE To analyze pathobiology of ischiocavernosus (IC) and bulbospongiosus (BS) muscles in long-term diabetic male rats and its implication on erectile dysfunction (ED). METHODS Male rats were grouped into control and diabetic rats (received single injection of 60 mg/kg bw. of streptozotocin [STZ]). At 120th day, the animals were subjected to various analyses like serum hormone, penile reflex, electromyography of IC and BS muscles, after euthanasia IC and BS muscles were processed for morphological, histology, histometric analysis, immunostaining and immunoblotting synaptophysin, nNOS and NADPH diaphorase histochemistry. RESULTS Significant reduction in serum hormone level, penile reflex, reduced action potential or activity in both these muscles and wide range of histological alterations were observed in STZ rats. Muscles showed significant reduction in the diameter, volume and numerical density of the fiber in both muscles of STZ rats. Synaptophysin, nNOS and NADPH diaphorase were significantly reduced in diabetic animal IC and BS. CONCLUSION Severe neuromuscular circuitry alteration in IC and BS. Study concludes that degenerative changes in IC and BS may play a major role in ED in diabetic condition. Indicating diabetic-induced postsynaptic neuronal degeneration along with impaired motor action of the muscle and severe muscle degeneration affecting ED.
Collapse
Affiliation(s)
- Prakash Seppan
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, Tamil Nadu, India
| | - Ibrahim Muhammed
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, Tamil Nadu, India
| | - Zafar Iqbal Khan Mohammad
- Department of Anatomy, Dr. Arcot Lakshmanasamy Mudaliar Postgraduate Institute of Basic Medical Sciences, University of Madras, Chennai, Tamil Nadu, India
| | - Sathya Bharathy Sathyanathan
- Department of Electrical and Electronics Engineering, Loyola-ICAM College of Engineering and Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
30
|
Daiber A, Chlopicki S. Revisiting pharmacology of oxidative stress and endothelial dysfunction in cardiovascular disease: Evidence for redox-based therapies. Free Radic Biol Med 2020; 157:15-37. [PMID: 32131026 DOI: 10.1016/j.freeradbiomed.2020.02.026] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 02/05/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
According to the latest Global Burden of Disease Study data, non-communicable diseases in general and cardiovascular disease (CVD) in particular are the leading cause of premature death and reduced quality of life. Demographic shifts, unhealthy lifestyles and a higher burden of adverse environmental factors provide an explanation for these findings. The expected growing prevalence of CVD requires enhanced research efforts for identification and characterisation of novel therapeutic targets and strategies. Cardiovascular risk factors including classical (e.g. hypertension, diabetes, hypercholesterolaemia) and non-classical (e.g. environmental stress) factors induce the development of endothelial dysfunction, which is closely associated with oxidant stress and vascular inflammation and results in CVD, particularly in older adults. Most classically successful therapies for CVD display vasoprotective, antioxidant and anti-inflammatory effects, but were originally designed with other therapeutic aims. So far, only a few 'redox drugs' are in clinical use and many antioxidant strategies have not met expectations. With the present review, we summarise the actual knowledge on CVD pathomechanisms, with special emphasis on endothelial dysfunction, adverse redox signalling and oxidative stress, highlighting the preclinical and clinical evidence. In addition, we provide a brief overview of established CVD therapies and their relation to endothelial dysfunction and oxidative stress. Finally, we discuss novel strategies for redox-based CVD therapies trying to explain why, despite a clear link between endothelial dysfunction and adverse redox signalling and oxidative stress, redox- and oxidative stress-based therapies have not yet provided a breakthrough in the treatment of endothelial dysfunction and CVD.
Collapse
Affiliation(s)
- Andreas Daiber
- The Center for Cardiology, Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Langenbeckstr. 1, 55131, Mainz, Germany; The Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Stefan Chlopicki
- The Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Bobrzynskiego 14, 30-348, Krakow, Poland; Jagiellonian University Medical College, Grzegorzecka 16, 31-531, Krakow, Poland.
| |
Collapse
|
31
|
Akanji MA, Adeyanju AA, Rotimi D, Adeyemi OS. Nitric Oxide Balance in Health and Diseases: Implications for New Treatment Strategies. Open Biochem J 2020. [DOI: 10.2174/1874091x02014010025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Nitric Oxide (NO) is an essential signaling molecule with diverse physiological functions in humans. The steady-state concentration and site of production of nitric oxide determine its effects in biological systems. The human cells are exposed to both beneficial and harmful effects of NO. These dual effects of NO could depend on its local concentration in the cells. Additionally, the rate of synthesis, translocation, direct interaction with other molecules, and signals contribute to the biochemical and physiological effects of NO. In this review, the biochemical and physiological role of NO, particularly in health and disease as touching on cell signaling, oxidative stress, immunity, as well as cardiovascular protection amongst others, is focused on. Therefore, this review objectively discusses the dual functionality of NO in living cells.
Collapse
|
32
|
Endothelial Dysfunction: A Contributor to Adverse Cardiovascular Remodeling and Heart Failure Development in Type 2 Diabetes beyond Accelerated Atherogenesis. J Clin Med 2020; 9:jcm9072090. [PMID: 32635218 PMCID: PMC7408687 DOI: 10.3390/jcm9072090] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/30/2020] [Accepted: 06/30/2020] [Indexed: 12/12/2022] Open
Abstract
Endothelial dysfunction, associated with depressed nitric oxide (NO) bioavailability, is awell-recognized contributor to both accelerated atherogenesis and microvascular complications intype 2 diabetes (DM). However, growing evidence points to the comorbidities-driven endothelialdysfunction within coronary microvessels as a key player responsible for left ventricular (LV)diastolic dysfunction, restrictive LV remodeling and heart failure with preserved ejection fraction(HFpEF), the most common form of heart failure in DM. In this review we have described: (1)multiple cellular pathways which may link depressed NO bioavailability to LV diastolicdysfunction and hypertrophy; (2) hemodynamic consequences and prognostic effects of restrictiveLV remodeling and combined diastolic and mild systolic LV dysfunction on cardiovascularoutcomes in DM and HFpEF, with a focus on the clinical relevance of endothelial dysfunction; (3)novel therapeutic strategies to improve endothelial function in DM. In summary, beyondassociations with accelerated atherogenesis and microvascular complications, endothelialdysfunction supplements the multiple interwoven pathways affecting cardiomyocytes, endothelialcells and the extracellular matrix with consequent LV dysfunction in DM patients. The associationamongst impaired endothelial function, reduced coronary flow reserve, combined LV diastolic anddiscrete systolic dysfunction, and low LV stroke volume and preload reserve-all of which areadverse outcome predictors-is a dangerous constellation of inter-related abnormalities, underlyingthe development of heart failure. Nevertheless, the relevance of endothelial effects of novel drugsin terms of their ability to attenuate cardiovascular remodeling and delay heart failure onset in DMpatients remains to be investigated.
Collapse
|
33
|
Regulation of Vascular Function and Inflammation via Cross Talk of Reactive Oxygen and Nitrogen Species from Mitochondria or NADPH Oxidase-Implications for Diabetes Progression. Int J Mol Sci 2020; 21:ijms21103405. [PMID: 32408480 PMCID: PMC7279344 DOI: 10.3390/ijms21103405] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays a key role for the development of cardiovascular, metabolic, and neurodegenerative disease. This concept has been proven by using the approach of genetic deletion of reactive oxygen and nitrogen species (RONS) producing, pro-oxidant enzymes as well as by the overexpression of RONS detoxifying, antioxidant enzymes leading to an amelioration of the severity of diseases. Vice versa, the development and progression of cardiovascular diseases is aggravated by overexpression of RONS producing enzymes as well as deletion of RONS detoxifying enzymes. We have previously identified cross talk mechanisms between different sources of RONS, which can amplify the oxidative stress-mediated damage. Here, the pathways and potential mechanisms leading to this cross talk are analyzed in detail and highlighted by selected examples from the current literature and own data including hypoxia, angiotensin II (AT-II)-induced hypertension, nitrate tolerance, aging, and others. The general concept of redox-based activation of RONS sources via “kindling radicals” and enzyme-specific “redox switches” as well as the interaction with redox-sensitive inflammatory pathways are discussed. Here, we present evidence for the existence of such cross talk mechanisms in the setting of diabetes and critically assess their contribution to the severity of diabetic complications.
Collapse
|
34
|
Daiber A, Kröller-Schön S, Oelze M, Hahad O, Li H, Schulz R, Steven S, Münzel T. Oxidative stress and inflammation contribute to traffic noise-induced vascular and cerebral dysfunction via uncoupling of nitric oxide synthases. Redox Biol 2020; 34:101506. [PMID: 32371009 PMCID: PMC7327966 DOI: 10.1016/j.redox.2020.101506] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
Environmental pollution and non-chemical stressors such as mental stress or traffic noise exposure are increasingly accepted as health risk factors with substantial contribution to chronic noncommunicable diseases (e.g. cardiovascular, metabolic and mental). Whereas the mechanisms of air pollution-mediated adverse health effects are well characterized, the mechanisms of traffic noise exposure are not completely understood, despite convincing clinical and epidemiological evidence for a significant contribution of environmental noise to overall mortality and disability. The initial mechanism of noise-induced cardiovascular, metabolic and mental disease is well defined by the „noise reaction model“ and consists of neuronal activation involving the hypothalamic-pituitary-adrenal (HPA) axis as well as the sympathetic nervous system, followed by a classical stress response via cortisol and catecholamines. Stress pathways are initiated by noise-induced annoyance and sleep deprivation/fragmentation. This review highlights the down-stream pathophysiology of noise-induced mental stress, which is based on an induction of inflammation and oxidative stress. We highlight the sources of reactive oxygen species (ROS) involved and the known targets for noise-induced oxidative damage. Part of the review emphasizes noise-triggered uncoupling/dysregulation of endothelial and neuronal nitric oxide synthase (eNOS and nNOS) and its central role for vascular dysfunction. Exposure to (traffic) noise causes non-auditory (indirect) cardiovascular and cerebral health harms via neuronal activation. Noise activates the HPA axis and sympathetic nervous system increasing levels of stress hormones, vasoconstrictors and ROS. Noise induces inflammation and stimulates several ROS sources leading to cerebral and cardiovascular oxidative damage. Noise leads to eNOS and nNOS uncoupling contributing to cardiometabolic disease and cognitive impairment.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| | - Swenja Kröller-Schön
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Matthias Oelze
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Omar Hahad
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Mainz, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Sebastian Steven
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany
| | - Thomas Münzel
- Center for Cardiology, Molecular Cardiology, University Medical Center, Mainz, Germany; Partner Site Rhine-Main, German Center for Cardiovascular Research (DZHK), Langenbeckstr. 1, 55131, Mainz, Germany.
| |
Collapse
|
35
|
Kraft VN, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C, Zandkarimi F, Merl-Pham J, Bao X, Anastasov N, Kössl J, Brandner S, Daniels JD, Schmitt-Kopplin P, Hauck SM, Stockwell BR, Hadian K, Schick JA. GTP Cyclohydrolase 1/Tetrahydrobiopterin Counteract Ferroptosis through Lipid Remodeling. ACS CENTRAL SCIENCE 2020; 6:41-53. [PMID: 31989025 PMCID: PMC6978838 DOI: 10.1021/acscentsci.9b01063] [Citation(s) in RCA: 740] [Impact Index Per Article: 148.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Indexed: 05/03/2023]
Abstract
Ferroptosis is an iron-dependent form of regulated cell death linking iron, lipid, and glutathione levels to degenerative processes and tumor suppression. By performing a genome-wide activation screen, we identified a cohort of genes antagonizing ferroptotic cell death, including GTP cyclohydrolase-1 (GCH1) and its metabolic derivatives tetrahydrobiopterin/dihydrobiopterin (BH4/BH2). Synthesis of BH4/BH2 by GCH1-expressing cells caused lipid remodeling, suppressing ferroptosis by selectively preventing depletion of phospholipids with two polyunsaturated fatty acyl tails. GCH1 expression level in cancer cell lines stratified susceptibility to ferroptosis, in accordance with its expression in human tumor samples. The GCH1-BH4-phospholipid axis acts as a master regulator of ferroptosis resistance, controlling endogenous production of the antioxidant BH4, abundance of CoQ10, and peroxidation of unusual phospholipids with two polyunsaturated fatty acyl tails. This demonstrates a unique mechanism of ferroptosis protection that is independent of the GPX4/glutathione system.
Collapse
Affiliation(s)
- Vanessa
A. N. Kraft
- Institute
of Molecular Toxicology and Pharmacology, Genetics and Cellular Engineering
Group, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Carla T. Bezjian
- Department
of Chemistry, Columbia University, 550 West 120th Street, MC4846, New York, New York 10027, United States
| | - Susanne Pfeiffer
- Institute
of Molecular Toxicology and Pharmacology, Genetics and Cellular Engineering
Group, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Larissa Ringelstetter
- Institute
of Molecular Toxicology and Pharmacology, Assay Development and Screening
Platform, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Constanze Müller
- Research
Unit Analytical BioGeoChemistry, HelmholtzZentrum
Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Fereshteh Zandkarimi
- Department
of Biological Sciences, Columbia University, New York, New York 10027, United States
| | - Juliane Merl-Pham
- Research
Unit Protein Science, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Xuanwen Bao
- Institute
of Radiation Biology, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Natasa Anastasov
- Institute
of Radiation Biology, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Johanna Kössl
- Institute
of Molecular Toxicology and Pharmacology, Genetics and Cellular Engineering
Group, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Stefanie Brandner
- Institute
of Molecular Toxicology and Pharmacology, Assay Development and Screening
Platform, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Jacob D. Daniels
- Department
of Pharmacology, Columbia University, New York, New York 10027, United States
| | - Philippe Schmitt-Kopplin
- Research
Unit Analytical BioGeoChemistry, HelmholtzZentrum
Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Stefanie M. Hauck
- Research
Unit Protein Science, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Brent R. Stockwell
- Department
of Chemistry, Columbia University, 550 West 120th Street, MC4846, New York, New York 10027, United States
- Department
of Biological Sciences, Columbia University, New York, New York 10027, United States
- E-mail:
| | - Kamyar Hadian
- Institute
of Molecular Toxicology and Pharmacology, Assay Development and Screening
Platform, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
- E-mail:
| | - Joel A. Schick
- Institute
of Molecular Toxicology and Pharmacology, Genetics and Cellular Engineering
Group, HelmholtzZentrum Muenchen, Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
- E-mail:
| |
Collapse
|
36
|
Lee I, Kim S, Nagar H, Choi SJ, Jeon BH, Piao S, Kim CS. CR6-interacting factor 1 deficiency reduces endothelial nitric oxide synthase activity by inhibiting biosynthesis of tetrahydrobiopterin. Sci Rep 2020; 10:842. [PMID: 31964986 PMCID: PMC6972730 DOI: 10.1038/s41598-020-57673-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 12/31/2019] [Indexed: 01/07/2023] Open
Abstract
Downregulation of CR6 interacting factor 1 (CRIF1) has been reported to induce mitochondrial dysfunction, resulting in reduced activity of endothelial nitric oxide synthase (eNOS) and NO production in endothelial cells. Tetrahydrobiopterin (BH4) is an important cofactor in regulating the balance between NO (eNOS coupling) and superoxide production (eNOS uncoupling). However, whether the decreased eNOS and NO production in CRIF1-deficient cells is associated with relative BH4 deficiency-induced eNOS uncoupling remains completely unknown. Our results showed that CRIF1 deficiency increased eNOS uncoupling and depleted levels of total biopterin and BH4 by reducing the enzymes of BH4 biosynthesis (GCH-1, PTS, SPR, and DHFR) in vivo and vitro, respectively. Supplementation of CRIF1-deficient cells with BH4 significantly increased the recovery of Akt and eNOS phosphorylation and NO synthesis. In addition, scavenging ROS with MitoTEMPO treatment replenished BH4 levels by elevating levels of GCH-1, PTS, and SPR, but with no effect on the level of DHFR. Downregulation of DHFR synthesis regulators p16 or p21 in CRIF1-deficient cells partially recovered the DHFR expression. In summary, CRIF1 deficiency inhibited BH4 biosynthesis and exacerbated eNOS uncoupling. This resulted in reduced NO production and increased oxidative stress, which contributes to endothelial dysfunction and is involved in the pathogenesis of cardiovascular diseases.
Collapse
Affiliation(s)
- Ikjun Lee
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Seonhee Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Harsha Nagar
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Su-Jeong Choi
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Byeong Hwa Jeon
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea
| | - Shuyu Piao
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea.
| | - Cuk-Seong Kim
- Department of Physiology & Medical Science, School of Medicine, Chungnam National University, Daejeon, 301-747, Republic of Korea.
| |
Collapse
|
37
|
Romão PVM, Palozi RAC, Guarnier LP, Silva AO, Lorençone BR, Nocchi SR, Moura CCDFS, Lourenço ELB, Silva DB, Gasparotto Junior A. Cardioprotective effects of Plinia cauliflora (Mart.) Kausel in a rabbit model of doxorubicin-induced heart failure. JOURNAL OF ETHNOPHARMACOLOGY 2019; 242:112042. [PMID: 31254629 DOI: 10.1016/j.jep.2019.112042] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 06/08/2019] [Accepted: 06/24/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In Brazil, the fruit of a native species that is popularly known as "jabuticaba" (Plinia cauliflora [Mart.] Kausel) is widely consumed fresh or used for the production of liqueur, juice, and jelly. In Brazilian folk medicine, this species is used to treat asthma, throat inflammation, and gastrointestinal and cardiovascular disturbances. However, no previous studies have reported its cardioprotective effects. AIM To evaluate the possible cardioprotective effects of a hydroethanolic extract of Plinia cauliflora (EEPC) in female rabbits in a model of doxorubicin-induced heart failure. MATERIAL AND METHODS EEPC was obtained and fractionated by solid phase extraction, and its constituents were determined by liquid chromatography coupled to diode array detector and mass spectrometry (LC-DAD-MS). Thirty female New Zealand rabbits received doxorubicin administration for 6 weeks to induce heart failure. EEPC was orally administered at doses of 75 and 150 mg/kg daily for 42 days. Enalapril (5 mg/kg) was used as a reference cardioprotective drug. At the end of the experimental period, blood pressure and heart rate were recorded. Serum parameters, including lipid profile, troponin, creatinine, nitrotyrosine, malondialdehyde, nitrite, and brain natriuretic peptide, were measured. The electrocardiographic profile and renal vascular reactivity were evaluated. Cardiac histopathology and ventricular morphometry were performed, and the tissue enzymatic antioxidant system was investigated. RESULTS A total of 37 compounds were detected in EEPC, including organic acids, phenolic acid derivatives, flavonoids, anthocyanins, and hydrolysable tannins (gallotannins and ellagitannins). EEPC treatment induced a cardiorenal protective response, prevented hemodynamic and functional alterations, and prevented ventricle remodeling. These effects were associated with the normalization of creatinine and brain natriuretic peptide levels and modulation of the tecidual antioxidant defense system. CONCLUSION The present study demonstrated that EEPC may prevent doxorubicin-induced heart failure by modulating the antioxidant defense system, reducing reactive oxygen species-induced damage, preventing alterations of hemodynamic and endothelial function, and preventing damage to the cardiac structure. EEPC, especially at the highest dose tested, may be considered a cardioprotective coadjuvant to prevent doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Paulo Vitor Moreira Romão
- Laboratório de Eletrofisiologia e Farmacologia Cardiovascular (LEFaC), Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Rhanany Alan Calloi Palozi
- Laboratório de Eletrofisiologia e Farmacologia Cardiovascular (LEFaC), Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Lucas Pires Guarnier
- Laboratório de Eletrofisiologia e Farmacologia Cardiovascular (LEFaC), Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Aniely Oliveira Silva
- Laboratório de Eletrofisiologia e Farmacologia Cardiovascular (LEFaC), Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Bethânia Rosa Lorençone
- Laboratório de Eletrofisiologia e Farmacologia Cardiovascular (LEFaC), Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil
| | - Samara Requena Nocchi
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | | | | | - Denise Brentan Silva
- Laboratório de Produtos Naturais e Espectrometria de Massas (LaPNEM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição (FACFAN), Universidade Federal do Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Arquimedes Gasparotto Junior
- Laboratório de Eletrofisiologia e Farmacologia Cardiovascular (LEFaC), Faculdade de Ciências da Saúde, Universidade Federal da Grande Dourados, Dourados, MS, Brazil.
| |
Collapse
|
38
|
Engineer A, Saiyin T, Greco ER, Feng Q. Say NO to ROS: Their Roles in Embryonic Heart Development and Pathogenesis of Congenital Heart Defects in Maternal Diabetes. Antioxidants (Basel) 2019; 8:antiox8100436. [PMID: 31581464 PMCID: PMC6826639 DOI: 10.3390/antiox8100436] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/09/2019] [Accepted: 09/18/2019] [Indexed: 12/12/2022] Open
Abstract
Congenital heart defects (CHDs) are the most prevalent and serious birth defect, occurring in 1% of all live births. Pregestational maternal diabetes is a known risk factor for the development of CHDs, elevating the risk in the child by more than four-fold. As the prevalence of diabetes rapidly rises among women of childbearing age, there is a need to investigate the mechanisms and potential preventative strategies for these defects. In experimental animal models of pregestational diabetes induced-CHDs, upwards of 50% of offspring display congenital malformations of the heart, including septal, valvular, and outflow tract defects. Specifically, the imbalance of nitric oxide (NO) and reactive oxygen species (ROS) signaling is a major driver of the development of CHDs in offspring of mice with pregestational diabetes. NO from endothelial nitric oxide synthase (eNOS) is crucial to cardiogenesis, regulating various cellular and molecular processes. In fact, deficiency in eNOS results in CHDs and coronary artery malformation. Embryonic hearts from diabetic dams exhibit eNOS uncoupling and oxidative stress. Maternal treatment with sapropterin, a cofactor of eNOS, and antioxidants such as N-acetylcysteine, vitamin E, and glutathione as well as maternal exercise have been shown to improve eNOS function, reduce oxidative stress, and lower the incidence CHDs in the offspring of mice with pregestational diabetes. This review summarizes recent data on pregestational diabetes-induced CHDs, and offers insights into the important roles of NO and ROS in embryonic heart development and pathogenesis of CHDs in maternal diabetes.
Collapse
Affiliation(s)
- Anish Engineer
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, ON, N6A 5C1, Canada.
| | - Tana Saiyin
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, ON, N6A 5C1, Canada.
| | - Elizabeth R Greco
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, ON, N6A 5C1, Canada.
| | - Qingping Feng
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, Ontario, ON, N6A 5C1, Canada.
| |
Collapse
|
39
|
Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7092151. [PMID: 31341533 PMCID: PMC6612399 DOI: 10.1155/2019/7092151] [Citation(s) in RCA: 406] [Impact Index Per Article: 67.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/20/2019] [Indexed: 02/08/2023]
Abstract
Cardiovascular disease is a leading cause of death and reduced quality of life, proven by the latest data of the Global Burden of Disease Study, and is only gaining in prevalence worldwide. Clinical trials have identified chronic inflammatory disorders as cardiovascular risks, and recent research has revealed a contribution by various inflammatory cells to vascular oxidative stress. Atherosclerosis and cardiovascular disease are closely associated with inflammation, probably due to the close interaction of inflammation with oxidative stress. Classical therapies for inflammatory disorders have demonstrated protective effects in various models of cardiovascular disease; especially established drugs with pleiotropic immunomodulatory properties have proven beneficial cardiovascular effects; normalization of oxidative stress seems to be a common feature of these therapies. The close link between inflammation and redox balance was also supported by reports on aggravated inflammatory phenotype in the absence of antioxidant defense proteins (e.g., superoxide dismutases, heme oxygenase-1, and glutathione peroxidases) or overexpression of reactive oxygen species producing enzymes (e.g., NADPH oxidases). The value of immunomodulation for the treatment of cardiovascular disease was recently supported by large-scale clinical trials demonstrating reduced cardiovascular mortality in patients with established atherosclerotic disease when treated by highly specific anti-inflammatory therapies (e.g., using monoclonal antibodies against cytokines). Modern antidiabetic cardiovascular drugs (e.g., SGLT2 inhibitors, DPP-4 inhibitors, and GLP-1 analogs) seem to share these immunomodulatory properties and display potent antioxidant effects, all of which may explain their successful lowering of cardiovascular risk.
Collapse
|
40
|
Li Y, Deng SL, Lian ZX, Yu K. Roles of Toll-Like Receptors in Nitroxidative Stress in Mammals. Cells 2019; 8:cells8060576. [PMID: 31212769 PMCID: PMC6627996 DOI: 10.3390/cells8060576] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 06/08/2019] [Accepted: 06/10/2019] [Indexed: 01/20/2023] Open
Abstract
Free radicals are important antimicrobial effectors that cause damage to DNA, membrane lipids, and proteins. Professional phagocytes produce reactive oxygen species (ROS) and reactive nitrogen species (RNS) that contribute towards the destruction of pathogens. Toll-like receptors (TLRs) play a fundamental role in the innate immune response and respond to conserved microbial products and endogenous molecules resulting from cellular damage to elicit an effective defense against invading pathogens, tissue injury, or cancer. In recent years, several studies have focused on how the TLR-mediated activation of innate immune cells leads to the production of pro-inflammatory factors upon pathogen invasion. Here, we review recent findings that indicate that TLRs trigger a signaling cascade that induces the production of reactive oxygen and nitrogen species.
Collapse
Affiliation(s)
- Yao Li
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Shou-Long Deng
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Zheng-Xing Lian
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| | - Kun Yu
- Beijing Key Laboratory for Animal Genetic Improvement, National Engineering Laboratory for Animal Breeding, Key Laboratory of Animal Genetics and Breeding of the Ministry of Agriculture, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China.
| |
Collapse
|
41
|
Pharmacological inhibition of guanosine triphosphate cyclohydrolase1 elevates tyrosine phosphorylation of caveolin1 and cellular senescence. Eur J Pharmacol 2019; 848:1-10. [PMID: 30690003 DOI: 10.1016/j.ejphar.2019.01.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/15/2019] [Accepted: 01/18/2019] [Indexed: 01/02/2023]
Abstract
The role of 2,4-diamino-6-hydroxypyrimidine (DAHP), on cellular-senescence remains unclear as differential effects of DAHP have been reported in cardiovascular and cerebrovascular systems. We investigated the effect of pharmacologically-induced guanosine-triphosphate-cyclohydrolase1 (GTPCH1)-inhibition, through DAHP, on cellular-senescence in experimentally-induced diabetic and non-diabetic Wistar rats. Cellular-senescence was evaluated through senescence-associated events, namely, cell-cycle-arrest of peripheral blood mononuclear cells (PBMNCs); myocardial DNA fragmentation, total antioxidant capacity (TAC), telomerase-activity, nicotinamide adenine dinucleotide (NAD+)-content and tyrosine14-phosphorylation of caveolin1 (pY14) in similarly-aged, pubertal Wistar rats with streptozotocin (STZ) and/or DAHP. Oxidative stress (OS) indices such as myocardial biopterin concentrations (tetrahydrobiopterin-BH4 and dihydrobiopterin-BH2) and plasma total nitrite and nitrate (NOx) were determined. DAHP, per se, exhibited distinct senescence; in addition, in STZ+DAHP (the cardiomyopathy model), there was a marked accumulation of cells in G0G1 phase, as evidenced through flow-cytometry analysis, as-well-as fragmented DNA, than the respective controls suggesting the DAHP-mediated onset of senescence in circulating cells and the myocardium, with or without STZ. Concentrations of BH4 and BH2, and NOx were impaired in STZ and/or DAHP, indicating elevated OS in the treatment groups. In the independent treatment groups or the combination treatment, typical senescence indicators including myocardial telomerase-activity, NAD+-content and TAC were significantly reduced, while there was a marked elevation in the concentrations of pY14 as compared to the respective controls, reinforcing the occurrence of senescence in PBMNCs and the myocardium. We postulate that DAHP promotes early onset of cellular-senescence, potentially through OS-mediated cellular events in diabetic or non-diabetic models.
Collapse
|
42
|
|
43
|
Perhal A, Wolf S, Jamous YF, Langer A, Abd Alla J, Quitterer U. Increased Reactive Oxygen Species Generation Contributes to the Atherogenic Activity of the B2 Bradykinin Receptor. Front Med (Lausanne) 2019; 6:32. [PMID: 30847343 PMCID: PMC6393342 DOI: 10.3389/fmed.2019.00032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 02/04/2019] [Indexed: 11/21/2022] Open
Abstract
Atherosclerosis and ensuing cardiovascular disease are major causes of death with insufficient treatment options. In search for pathomechanisms of atherosclerosis, we investigated the impact of the B2 bradykinin receptor, Bdkrb2, on atherosclerotic lesion formation, because to date it is not clear whether the B2 bradykinin receptor is atheroprotective or atherogenic. As a model of atherosclerosis, we used hypercholesterolemic ApoE-deficient (apolipoprotein E-deficient) mice, which develop atherosclerotic lesions in the aorta with increasing age. The role of Bdkrb2 in atherosclerosis was studied in ApoE-deficient mice, which were either Bdkrb2-deficient, or had moderately increased aortic B2 bradykinin receptor protein levels induced by transgenic BDKRB2 expression under control of the ubiquitous CMV promoter. We found that Bdkrb2 deficiency led to a significantly decreased atherosclerotic plaque area whereas transgenic BDKRB2 expression enhanced atherosclerotic lesion formation in the aorta of ApoE-deficient mice at an age of 8 months. Concomitantly, the aortic content of reactive oxygen species (ROS) was higher in BDKRB2-expressing mice whereas Bdkrb2 deficiency decreased aortic ROS levels of ApoE-deficient mice. In addition, aortic nitrate as a marker of nitric oxide activity and the endothelial nitric oxide synthase (eNOS) co-factor, tetrahydrobiopterin (BH4) were reduced in BDKRB2-expressing ApoE-deficient mice. The decreased aortic BH4 content could be a consequence of increased ROS generation and down-regulated aortic expression of the BH4-synthesizing enzyme, Gch1 (GTP cyclohydrolase 1). In agreement with a causal involvement of decreased BH4 levels in the atherogenic function of BDKRB2, we found that treatment with the BH4 analog, sapropterin, significantly retarded atherosclerotic plaque formation in BDKRB2-expressing ApoE-deficient mice. Together our data show that the B2 bradykinin receptor is atherogenic, and the atherosclerosis-promoting function of BDKRB2 is partially caused by decreased aortic BH4 levels, which could account for eNOS uncoupling and further enhancement of ROS generation.
Collapse
Affiliation(s)
- Alexander Perhal
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Stefan Wolf
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Yahya F Jamous
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Andreas Langer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Joshua Abd Alla
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Ursula Quitterer
- Molecular Pharmacology, Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland.,Department of Medicine, Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
44
|
Zhu ML, Wang G, Wang H, Guo YM, Song P, Xu J, Li P, Wang S, Yang L. Amorphous nano-selenium quantum dots improve endothelial dysfunction in rats and prevent atherosclerosis in mice through Na +/H + exchanger 1 inhibition. Vascul Pharmacol 2019; 115:26-32. [PMID: 30695730 DOI: 10.1016/j.vph.2019.01.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/08/2019] [Accepted: 01/25/2019] [Indexed: 12/27/2022]
Abstract
AIM Selenium, a trace element involved in important enzymatic activities inside the body, has protective effects against cardiovascular diseases including atherosclerosis. The safe dose of selenium in the organism is very narrow, limiting the supplementation of selenium in diet. The aim of this study is to explore whether selenium quantum dots (SeQDs) prevent atherosclerosis and to investigate the potential mechanisms. METHODS An amorphous form of SeQDs (A-SeQDs) and a crystalline form of SeQDs (C-SeQDs) were prepared through self-redox decomposition of selenosulfate precursor. Endothelial dysfunction was induced by balloon injury plus high fat diet (HFD) in rats. Atherosclerotic model was established by feeding Apoe-/- mice with HFD. RESULTS Administrations of A-SeQDs but not C-SeQDs dramatically improved endothelium-dependent relaxation, and accelerated would healing in primary endothelial cells isolated from rats, which was comprised by co-treatment of LiCl. Lentivirus-mediated knockdown of Na+/H+ exchanger 1 (NHE1) abolished LiCl-induced endothelial dysfunction in rats. In cultured endothelial cells, A-SeQDs, as well as cariporide, inhibited NHE1 activities, decreased intracellular pH value and Ca2+ concentration, and reduced calpain activity increased by ox-LDL. These protective effects of A-SeQDs were reversed by LiCl treatment in endothelial cells. In Apoe-/- mice feeding with HFD, A-SeQDs prevented endothelial dysfunction and reduced the size of atherosclerotic plaque in aortic arteries. Further, lentivirus-mediated NHE1 gene overexpression abolished the protective effects of A-SeQDs against endothelial dysfunction and atherosclerosis in Apoe-/- mice. CONCLUSION A-SeQDs prevents endothelial dysfunction and the growth of atherosclerotic plaque through NHE1 inhibition and subsequent inactivation of Ca2+/calpain signaling. Clinically, the administration of A-SeQDs is an effective approach to treat atherosclerosis.
Collapse
Affiliation(s)
- Mo-Li Zhu
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China; School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Ge Wang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China; School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - He Wang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China
| | - Yu-Ming Guo
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China
| | - Ping Song
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Jian Xu
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China
| | - Peng Li
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Shuangxi Wang
- School of Pharmacy, Xinxiang Medical University, Xinxiang, Henan, China.
| | - Lin Yang
- Collaborative Innovation Center of Henan Province for Green Manufacturing of Fine Chemicals, Key Laboratory of Green Chemical Media and Reactions, Ministry of Education, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan, China.
| |
Collapse
|
45
|
Daiber A, Xia N, Steven S, Oelze M, Hanf A, Kröller-Schön S, Münzel T, Li H. New Therapeutic Implications of Endothelial Nitric Oxide Synthase (eNOS) Function/Dysfunction in Cardiovascular Disease. Int J Mol Sci 2019; 20:ijms20010187. [PMID: 30621010 PMCID: PMC6337296 DOI: 10.3390/ijms20010187] [Citation(s) in RCA: 148] [Impact Index Per Article: 24.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 12/27/2018] [Accepted: 12/28/2018] [Indexed: 02/07/2023] Open
Abstract
The Global Burden of Disease Study identified cardiovascular risk factors as leading causes of global deaths and life years lost. Endothelial dysfunction represents a pathomechanism that is associated with most of these risk factors and stressors, and represents an early (subclinical) marker/predictor of atherosclerosis. Oxidative stress is a trigger of endothelial dysfunction and it is a hall-mark of cardiovascular diseases and of the risk factors/stressors that are responsible for their initiation. Endothelial function is largely based on endothelial nitric oxide synthase (eNOS) function and activity. Likewise, oxidative stress can lead to the loss of eNOS activity or even “uncoupling” of the enzyme by adverse regulation of well-defined “redox switches” in eNOS itself or up-/down-stream signaling molecules. Of note, not only eNOS function and activity in the endothelium are essential for vascular integrity and homeostasis, but also eNOS in perivascular adipose tissue plays an important role for these processes. Accordingly, eNOS protein represents an attractive therapeutic target that, so far, was not pharmacologically exploited. With our present work, we want to provide an overview on recent advances and future therapeutic strategies that could be used to target eNOS activity and function in cardiovascular (and other) diseases, including life style changes and epigenetic modulations. We highlight the redox-regulatory mechanisms in eNOS function and up- and down-stream signaling pathways (e.g., tetrahydrobiopterin metabolism and soluble guanylyl cyclase/cGMP pathway) and their potential pharmacological exploitation.
Collapse
Affiliation(s)
- Andreas Daiber
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany.
| | - Ning Xia
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Sebastian Steven
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Matthias Oelze
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Alina Hanf
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Swenja Kröller-Schön
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| | - Thomas Münzel
- Center for Cardiology, Cardiology I-Laboratory of Molecular Cardiology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131 Mainz, Germany.
| | - Huige Li
- Department of Pharmacology, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany.
| |
Collapse
|
46
|
Natarajan M, Habib SL, Reddick RL, Delma CR, Manickam K, Prihoda TJ, Werner SL, Mohan S. Endothelial cell-specific overexpression of endothelial nitric oxide synthase in Ins2Akita mice exacerbates diabetic nephropathy. J Diabetes Complications 2019; 33:23-32. [PMID: 30424931 PMCID: PMC6344355 DOI: 10.1016/j.jdiacomp.2018.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 08/13/2018] [Accepted: 10/07/2018] [Indexed: 10/28/2022]
Abstract
Previous studies demonstrated that global deficiency of eNOS in diabetic mice exacerbated renal lesions and that overexpression of eNOS may protect against tissue injury. Our study revealed for the first time overexpression of eNOS leads to disease progression rather than protection. Transgenic mice selectively expressing eNOS in endothelial cells (eNOSTg) were cross bred with Ins2Akita type-1 (AK) diabetic mice to generate eNOS overexpressing eNOSTg/AK mice. Wild type, eNOSTg, AK and eNOSTg/AK mice were assessed for kidney function and blood glucose levels. Remarkably, overexpressing eNOSTg mice showed evidence of unpredicted glomerular injury with segmental mesangiolysis and occasional microaneurysms. Notably, in eNOSTg/AK mice overexpression of eNOS led to increased glomerular/endothelial injury that was associated with increased superoxide levels and renal dysfunction. Results indicate for the first time that overexpressing eNOS in endothelial cells cannot ameliorate diabetic lesions, but paradoxically leads to progression of nephropathy likely due to eNOS uncoupling and superoxide upsurge. This novel finding has a significant impact on current therapeutic strategies to improve endothelial function and prevent progression of diabetic renal disease. Further, the eNOSTg/AK model developed in this study has significant translational potentials for elucidating the underlying mechanism implicated in the deflected function of eNOS in diabetic nephropathy.
Collapse
Affiliation(s)
- Mohan Natarajan
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Samy L Habib
- Geriatric Research Education and Clinical Center, South Texas Veterans Healthcare System and Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Robert L Reddick
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Caroline R Delma
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Krishnan Manickam
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Thomas J Prihoda
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sherry L Werner
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sumathy Mohan
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA.
| |
Collapse
|
47
|
Knapp M, Tu X, Wu R. Vascular endothelial dysfunction, a major mediator in diabetic cardiomyopathy. Acta Pharmacol Sin 2019; 40:1-8. [PMID: 29867137 PMCID: PMC6318313 DOI: 10.1038/s41401-018-0042-6] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2018] [Accepted: 04/06/2018] [Indexed: 12/23/2022] Open
Abstract
Diabetes mellitus is currently a major public health problem. A common complication of diabetes is cardiac dysfunction, which is recognized as a microvascular disease that leads to morbidity and mortality in diabetic patients. While ischemic events are commonly observed in diabetic patients, the risk for developing heart failure is also increased, independent of the severity of coronary artery disease and hypertension. This diabetes-associated clinical entity is considered a distinct disease process referred to as "diabetic cardiomyopathy". However, it is not clear how diabetes promotes cardiac dysfunction. Vascular endothelial dysfunction is thought to be one of the key risk factors. The impact of diabetes on the endothelium involves several alterations, including hyperglycemia, fatty acid oxidation, reduced nitric oxide (NO), oxidative stress, inflammatory activation, and altered barrier function. The current review provides an update on mechanisms that specifically target endothelial dysfunction, which may lead to diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Maura Knapp
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA
| | - Xin Tu
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA
| | - Rongxue Wu
- Department of Medicine, Section of Cardiology, University of Chicago, Chicago, USA.
| |
Collapse
|
48
|
Yu L, Liu H. Perillaldehyde prevents the formations of atherosclerotic plaques through recoupling endothelial nitric oxide synthase. J Cell Biochem 2018; 119:10204-10215. [DOI: 10.1002/jcb.27362] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 06/26/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Li Yu
- Department of Physiology, School of Basic Medical Sciences Jinzhou Medical University Jinzhou China
- Institue of Eyes Jinzhou Medical University Jinzhou China
| | - Hua Liu
- Institue of Eyes Jinzhou Medical University Jinzhou China
| |
Collapse
|
49
|
Sagoo MK, Gnudi L. Diabetic nephropathy: Is there a role for oxidative stress? Free Radic Biol Med 2018; 116:50-63. [PMID: 29305106 DOI: 10.1016/j.freeradbiomed.2017.12.040] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/27/2017] [Accepted: 12/31/2017] [Indexed: 01/06/2023]
Abstract
Oxidative stress has been implicated in the pathophysiology of diabetic nephropathy. Studies in experimental animal models of diabetes strongly implicate oxidant species as a major determinant in the pathophysiology of diabetic kidney disease. The translation, in the clinical setting, of these concepts have been quite disappointing, and new theories have challenged the concepts that oxidative stress per se plays a role in the pathophysiology of diabetic kidney disease. The concept of mitochondrial hormesis has been introduced to explain this apparent disconnect. Hormesis is intended as any cellular process that exhibits a biphasic response to exposure to increasing amounts of a substance or condition: specifically, in diabetic kidney disease, oxidant species may represent, at determined concentration, an essential and potentially protective factor. It could be postulated that excessive production or inhibition of oxidant species formation might result in an adverse phenotype. This review discusses the evidence underlying these two apparent contradicting concepts, with the aim to propose and speculate on potential mechanisms underlying the role of oxidant species in the pathophysiology of diabetic nephropathy and possibly open future more efficient therapies to be tested in the clinical settings.
Collapse
Affiliation(s)
- Manpreet K Sagoo
- School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, UK
| | - Luigi Gnudi
- School of Cardiovascular Medicine & Sciences, British Heart Foundation Centre of Research Excellence, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, UK.
| |
Collapse
|
50
|
Abstract
Nitric oxide (NO) signalling has pleiotropic roles in biology and a crucial function in cardiovascular homeostasis. Tremendous knowledge has been accumulated on the mechanisms of the nitric oxide synthase (NOS)-NO pathway, but how this highly reactive, free radical gas signals to specific targets for precise regulation of cardiovascular function remains the focus of much intense research. In this Review, we summarize the updated paradigms on NOS regulation, NO interaction with reactive oxidant species in specific subcellular compartments, and downstream effects of NO in target cardiovascular tissues, while emphasizing the latest developments of molecular tools and biomarkers to modulate and monitor NO production and bioavailability.
Collapse
Affiliation(s)
- Charlotte Farah
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| |
Collapse
|