1
|
Doddaballapur DB, Heyes DJ, Miyan JA. Multiple Hits on Cerebral Folate, Tetrahydrobiopterin and Dopamine Metabolism in the Pathophysiology of Parkinson's Disorder: A Limited Study of Post-Mortem Human Brain Tissues. Metabolites 2025; 15:307. [PMID: 40422884 DOI: 10.3390/metabo15050307] [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: 03/14/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/28/2025] Open
Abstract
Background: Parkinson's disorder (PD) affects around 1:500 individuals and is associated with enlarged ventricles and symptoms of normal pressure hydrocephalus (NPH). These features suggest disrupted cerebrospinal fluid (CSF) dynamics and folate metabolism. With L-DOPA treatment showing diminishing benefits over time, there is an urgent need to investigate upstream metabolic disruptions, including folate and tetrahydrobiopterin (BH4) pathways, in post-mortem CSF and brain tissue to understand their roles in PD pathogenesis. Methods: CSF and brain tissue from 20 PD patients (mean age 84 years; 55% male; disease duration 10-30 years) and 20 controls (mean age 82 years; 50% male) were analysed. Western and Dot Blots measured proteins and metabolites, spectroscopic assays assessed enzyme activities, BH4 and Neopterin levels were measured using ELISA, and levels of hydrogen peroxide, used as a proxy for reactive oxygen species, and calcium were quantified using horseradish peroxidase and flame photometry assays, respectively. ClinVar genetic data were analysed for variants in genes encoding key enzymes. Statistical significance was assessed using unpaired t-tests (p < 0.05). Results: All enzymes were significantly reduced in PD compared to controls (p < 0.01) except for methyltetrahydrofolate reductase (MTHFR), which was elevated (p < 0.0001). Enzymes were functional in control but undetectable in PD CSF except tyrosine hydroxylase (TH). BH4 and Neopterin were elevated in PD CSF (p < 0.0001, p < 0.001) but significantly reduced (p < 0.001) or unchanged in tissue. Peroxide was increased in both PD CSF (p < 0.001) and tissue (p < 0.0001) selectively inhibiting TH. Calcium was 40% higher in PD than controls (p < 0.05). No pathogenic variants in enzyme genes were found in ClinVar data searches, suggesting the observed deficiencies are physiological. Conclusions: We identified significant disruptions in folate and BH4 pathways in PD, with enzyme deficiencies, oxidative stress and calcium dysregulation pointing to choroid plexus dysfunction. These findings highlight the choroid plexus and CSF as key players in cerebral metabolism and promote further exploration of these as therapeutic targets to address dopaminergic dysfunction and ventricular enlargement in PD.
Collapse
Affiliation(s)
- Dhruti Balakrishna Doddaballapur
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, 3.540 Stopford Building, Oxford Road, Manchester M13 9PT, UK
| | - Derren J Heyes
- Institute of Biotechnology, Faculty of Science & Engineering, The University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Jaleel A Miyan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, 3.540 Stopford Building, Oxford Road, Manchester M13 9PT, UK
| |
Collapse
|
2
|
Sampath C, Campbell M, Baptiste J, Carter J, Greene MS, Dhungana G, Chukkapalli S, Gangula PR. Polybacterial Periodontal Infection Alters oxidative and Inflammatory Biomarkers in Primary Human Aortic Endothelial Cell (pHAECs). JOURNAL OF PHARMACY AND PHARMACOLOGY RESEARCH 2025; 9:45-53. [PMID: 40370477 PMCID: PMC12077613 DOI: 10.26502/fjppr.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
Background Periodontitis (PD) is a highly prevalent inflammatory disease associated with complex microbial infection in the subgingival cavity. We have previously shown that polybacterial periodontal infection led to aortic atherosclerosis and lipid profile modulation; however, the underlying mechanism(s) has not been yet demonstrated. Methods Primary human aortic endothelial cells (pHAECs) were infected periodically for 48 hours either with P. gingivalis (monobacterial infection) or polybacterial periodontal pathogens, P. gingivalis (Pg), T. forsythia (Tf), T. denticola (Td), and F. nucleatum (Fn), using HoxBan coculture technique. Cell viability was assessed using MTT assay. Nitric oxide synthesis was measured in the form of total nitrite released into the media after incubation period. Inflammatory and oxidative gene expression were evaluated in the cell lysate using quantitative real-time polymerase chain reaction (qRT-PCR) at each time point (12-48h). Statistical analysis was performed using two-way ANOVA. Results pHAEC cell viability was significantly (p<0.05) reduced in both mono and poly-bacterial infection when compared with non-infected cells in a time dependent manner. Nitrite levels in the media were significantly impaired in both infection groups. mRNA expression for cytokines (TLR-4, IL-6, and TNF-α,) and inducible nitric oxide synthase (iNOS) were significantly (p<0.05) elevated in both experimental groups. In contrast, endothelial (e) NOS, tetrahydrobiopterin (BH4) synthesis, GCH -1, Nrf2, and Nqo 1 were significantly (p<0.05) reduced in both experimental groups. Finally, polybacterial infection group showed greater changes in cell viability, nitrite levels, cytokines, eNOS/BH4/Nrf2 expression in a time dependent manner. Conclusions Our study highlights the adverse effects of in vitro PD infection in NO mediated vascular endothelial cell function.
Collapse
Affiliation(s)
- Chethan Sampath
- Department of Oral Diagnostic Sciences & Research, Department of Periodontology, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| | - Michaela Campbell
- Department of Oral Diagnostic Sciences & Research, Department of Periodontology, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| | - Jonathan Baptiste
- Department of Oral Diagnostic Sciences & Research, Department of Periodontology, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| | - Jalah Carter
- Department of Oral Diagnostic Sciences & Research, Department of Periodontology, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| | - Mia-Sade Greene
- Department of Oral Diagnostic Sciences & Research, Department of Periodontology, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| | - Gunaraj Dhungana
- Department of Oral Diagnostic Sciences & Research, Department of Periodontology, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| | - Sasanka Chukkapalli
- Department of Biomedical Engineering, Texas A & M University, College Station, TX 77843, USA
| | - Pandu R Gangula
- Department of Oral Diagnostic Sciences & Research, Department of Periodontology, School of Dentistry, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
3
|
Kurhaluk N, Tkaczenko H. L-Arginine and Nitric Oxide in Vascular Regulation-Experimental Findings in the Context of Blood Donation. Nutrients 2025; 17:665. [PMID: 40004994 PMCID: PMC11858268 DOI: 10.3390/nu17040665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
This narrative review provides an analysis of the role of nitric oxide (NO) and its precursors, particularly L-arginine, in vascular regulation and health, with an emphasis on findings from our experimental research in animal models. NO serves as a critical mediator of vascular function, contributing to vasodilation, the regulation of blood flow, and the prevention of thrombosis. As a primary precursor of NO, L-arginine is essential for maintaining endothelial integrity, modulating mitochondrial function, and reducing oxidative damage. This review synthesises the data and contextualises these findings within the physiological challenges faced by blood donors, such as repeated blood donation and associated oxidative stress. It examines the effects of L-arginine supplementation on mitochondrial respiration, lipid peroxidation, and microsomal oxidation in different conditions, including differences in age, gender, and dietary interventions. The mechanisms by which L-arginine enhances NO production, improves vascular elasticity, and alleviates endothelial dysfunction caused by reduced NO bioavailability are also investigated. By integrating experimental findings with insights from the existing literature, this review provides a perspective on the potential of L-arginine supplementation to address the specific physiological needs of blood donors. It highlights the importance of personalised nutritional approaches in enhancing donor recovery and vascular resilience. In addition, this review assesses the wider implications of L-arginine supplementation in mitigating oxidative stress and preserving vascular function. The interplay between NO bioavailability, dietary factors, and physiological adaptation in blood donors is highlighted, along with the identification of current knowledge gaps and recommendations for future research. By presenting both original experimental evidence and a critical synthesis of the literature, this article highlights the therapeutic potential of NO precursors, particularly L-arginine, in promoting vascular health in the context of blood donation.
Collapse
Affiliation(s)
- Natalia Kurhaluk
- Institute of Biology, Pomeranian University in Słupsk, Arciszewski St. 22b, 76-200 Słupsk, Poland;
| | | |
Collapse
|
4
|
Pandian K, Huang L, Junaid A, Harms A, van Zonneveld AJ, Hankemeier T. Tracer-based metabolomics for profiling nitric oxide metabolites in a 3D microvessels-on-chip model. FASEB J 2024; 38:e70005. [PMID: 39171967 DOI: 10.1096/fj.202400553r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/19/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Endothelial dysfunction, prevalent in cardiovascular diseases (CVDs) and linked to conditions like diabetes, hypertension, obesity, renal failure, or hypercholesterolemia, is characterized by diminished nitric oxide (NO) bioavailability-a key signaling molecule for vascular homeostasis. Current two-dimensional (2D) in vitro studies on NO synthesis by endothelial cells (ECs) lack the crucial laminar shear stress, a vital factor in modulating the NO-generating enzyme, endothelial nitric oxide synthase (eNOS), under physiological conditions. Here we developed a tracer-based metabolomics approach to measure NO-specific metabolites with mass spectrometry (MS) and show the impact of fluid flow on metabolic parameters associated with NO synthesis using 2D and 3D platforms. Specifically, we tracked the conversion of stable-isotope labeled NO substrate L-Arginine to L-Citrulline and L-Ornithine to determine eNOS activity. We demonstrated clear responses in human coronary artery endothelial cells (HCAECs) cultured with 13C6, 15N4-L-Arginine, and treated with eNOS stimulator, eNOS inhibitor, and arginase inhibitor. Analysis of downstream metabolites, 13C6, 15N3 L-Citrulline and 13C5, 15N2 L-Ornithine, revealed distinct outcomes. Additionally, we evaluated the NO metabolic status in static 2D culture and 3D microvessel models with bidirectional and unidirectional fluid flow. Our 3D model exhibited significant effects, particularly in microvessels exposed to the eNOS stimulator, as indicated by the 13C6, 15N3 L-Citrulline/13C5, 15N2 L-Ornithine ratio, compared to the 2D culture. The obtained results indicate that the 2D static culture mimics an endothelial dysfunction status, while the 3D model with a unidirectional fluid flow provides a more representative physiological environment that provides a better model to study endothelial dysfunction.
Collapse
Affiliation(s)
- Kanchana Pandian
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Luojiao Huang
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Abidemi Junaid
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Amy Harms
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| | - Anton Jan van Zonneveld
- Department of Internal Medicine (Nephrology) and the Einthoven Laboratory for Vascular and Regenerative Medicine, Leiden University Medical Center (LUMC), Leiden, the Netherlands
| | - Thomas Hankemeier
- Division of Systems Biomedicine and Pharmacology, LACDR, Leiden University, Leiden, the Netherlands
| |
Collapse
|
5
|
Bulbule S, Gottschalk CG, Drosen ME, Peterson D, Arnold LA, Roy A. Dysregulation of tetrahydrobiopterin metabolism in myalgic encephalomyelitis/chronic fatigue syndrome by pentose phosphate pathway. J Cent Nerv Syst Dis 2024; 16:11795735241271675. [PMID: 39161795 PMCID: PMC11331476 DOI: 10.1177/11795735241271675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 06/24/2024] [Indexed: 08/21/2024] Open
Abstract
Background Tetrahydrobiopterin (BH4) and its oxidized derivative dihydrobiopterin (BH2) were found to be strongly elevated in ME/CFS patients with orthostatic intolerance (ME + OI). Objective However, the molecular mechanism of biopterin biogenesis is poorly understood in ME + OI subjects. Here, we report that the activation of the non-oxidative pentose phosphate pathway (PPP) plays a critical role in the biogenesis of biopterins (BH4 and BH2) in ME + OI subjects. Research Design and Results Microarray-based gene screening followed by real-time PCR-based validation, ELISA assay, and finally enzyme kinetic studies of glucose-6-phosphate dehydrogenase (G6PDH), transaldolase (TALDO1), and transketolase (TK) enzymes revealed that the augmentation of anaerobic PPP is critical in the regulations of biopterins. To further investigate, we devised a novel cell culture strategy to induce non-oxidative PPP by treating human microglial cells with ribose-5-phosphate (R5P) under a hypoxic condition of 85%N2/10%CO2/5%O2 followed by the analysis of biopterin metabolism via ELISA, immunoblot, and dual immunocytochemical analyses. Moreover, the siRNA knocking down of the taldo1 gene strongly inhibited the bioavailability of phosphoribosyl pyrophosphate (PRPP), reduced the expressions of purine biosynthetic enzymes, attenuated GTP cyclohydrolase 1 (GTPCH1), and suppressed subsequent production of BH4 and its metabolic conversion to BH2 in R5P-treated and hypoxia-induced C20 human microglia cells. These results confirmed that the activation of non-oxidative PPP is indeed required for the upregulation of both BH4 and BH2 via the purine biosynthetic pathway. To test the functional role of ME + OI plasma-derived biopterins, exogenously added plasma samples of ME + OI plasma with high BH4 upregulated inducible nitric oxide synthase (iNOS) and nitric oxide (NO) in human microglial cells indicating that the non-oxidative PPP-induced-biopterins could stimulate inflammatory response in ME + OI patients. Conclusion Taken together, our current research highlights that the induction of non-oxidative PPP regulates the biogenesis of biopterins contributing to ME/CFS pathogenesis.
Collapse
Affiliation(s)
- Sarojini Bulbule
- Research and Development Laboratory, Simmaron Research Institute, Milwaukee, WI, USA
| | - Carl Gunnar Gottschalk
- Research and Development Laboratory, Simmaron Research Institute, Milwaukee, WI, USA
- Simmaron Research Institute, Incline Village, NV, USA
| | - Molly E. Drosen
- Research and Development Laboratory, Simmaron Research Institute, Milwaukee, WI, USA
| | | | - Leggy A. Arnold
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| | - Avik Roy
- Research and Development Laboratory, Simmaron Research Institute, Milwaukee, WI, USA
- Simmaron Research Institute, Incline Village, NV, USA
- Department of Chemistry and Biochemistry and the Milwaukee Institute for Drug Discovery, University of Wisconsin-Milwaukee, Milwaukee, WI, USA
| |
Collapse
|
6
|
Kwakye-Nuako G, Middleton CE, McCall LI. Small molecule mediators of host-T. cruzi-environment interactions in Chagas disease. PLoS Pathog 2024; 20:e1012012. [PMID: 38457443 PMCID: PMC10923493 DOI: 10.1371/journal.ppat.1012012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/10/2024] Open
Abstract
Small molecules (less than 1,500 Da) include major biological signals that mediate host-pathogen-microbiome communication. They also include key intermediates of metabolism and critical cellular building blocks. Pathogens present with unique nutritional needs that restrict pathogen colonization or promote tissue damage. In parallel, parts of host metabolism are responsive to immune signaling and regulated by immune cascades. These interactions can trigger both adaptive and maladaptive metabolic changes in the host, with microbiome-derived signals also contributing to disease progression. In turn, targeting pathogen metabolic needs or maladaptive host metabolic changes is an important strategy to develop new treatments for infectious diseases. Trypanosoma cruzi is a single-celled eukaryotic pathogen and the causative agent of Chagas disease, a neglected tropical disease associated with cardiac and intestinal dysfunction. Here, we discuss the role of small molecules during T. cruzi infection in its vector and in the mammalian host. We integrate these findings to build a theoretical interpretation of how maladaptive metabolic changes drive Chagas disease and extrapolate on how these findings can guide drug development.
Collapse
Affiliation(s)
- Godwin Kwakye-Nuako
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Biomedical Sciences, School of Allied Health Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Caitlyn E. Middleton
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, United States of America
| | - Laura-Isobel McCall
- Department of Chemistry and Biochemistry, University of Oklahoma, Norman, Oklahoma, United States of America
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, California, United States of America
| |
Collapse
|
7
|
Abolfazli S, Mortazavi P, Kheirandish A, Butler AE, Jamialahmadi T, Sahebkar A. Regulatory effects of curcumin on nitric oxide signaling in the cardiovascular system. Nitric Oxide 2024; 143:16-28. [PMID: 38141926 DOI: 10.1016/j.niox.2023.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/25/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
The continuously rising prevalence of cardiovascular disease (CVD) globally substantially impacts the economic growth of developing countries. Indeed, one of the leading causes of death worldwide is unfavorable cardiovascular events. Reduced nitric oxide (NO) generation is the pathogenic foundation of endothelial dysfunction, which is regarded as the first stage in the development of a number of CVDs. Nitric oxide exerts an array of biological effects, including vasodilation, the suppression of vascular smooth muscle cell proliferation and the functional control of cardiac cells. Numerous treatment strategies aim to increase NO synthesis or upregulate downstream NO signaling pathways. The major component of Curcuma longa, curcumin, has long been utilized in traditional medicine to treat various illnesses, especially CVDs. Curcumin improves CV function as well as having important pleiotropic effects, such as anti-inflammatory and antioxidant, through its ability to increase the bioavailability of NO and to positively impact NO-related signaling pathways. In this review, we discuss the scientific literature relating to curcumin's positive effects on NO signaling and vascular endothelial function.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Parham Mortazavi
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Kheirandish
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Hemmat Highway, Tehran, Iran
| | - Alexandra E Butler
- Research Department, Royal College of Surgeons in Ireland, Bahrain, PO Box, 15503, Adliya, Bahrain
| | - Tannaz Jamialahmadi
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
8
|
Cai S, Huang S, Zhang W, Xiao H, Yu D, Zhong X, Tao P, Luo Y. Integrated bioinformatic analysis reveals NOS2 as a novel ferroptosis-related biomarker for pre-eclampsia. BMC Pregnancy Childbirth 2023; 23:719. [PMID: 37817070 PMCID: PMC10563238 DOI: 10.1186/s12884-023-06051-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 10/05/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Pre-eclampsia (PE) is a common condition in pregnancy; however, methods for early diagnosis and effective treatment options are lacking. Ferroptosis is a newly identified iron-dependent cell death pathway. The aim of this study was to investigate the role of ferroptosis-related genes in PE, the underlying mechanism, and their potential diagnostic value using a bioinformatics approach. METHODS We downloaded the GSE48424 and GSE98224 datasets from the Gene Expression Omnibus database. Differentially expressed genes (DEGs) between PE and healthy pregnancy samples were identified in the GSE48424 dataset and subjected to weighted gene co-expression network analysis; the most relevant modules were intersected with known ferroptosis-related genes to distinctly identify the role of ferroptosis in PE. We further searched transcription factors and microRNAs that are predicted to regulate these ferroptosis-related genes, and patients in the GSE48424 dataset were divided into two groups according to high or low expression of the key ferroptosis-related genes associated with PE. To obtain robust key ferroptosis-related genes in PE, we validated their expression levels in the external dataset GSE98224. Finally, the reverse transcription-quantitative polymerase chain reaction (RT-qPCR) assay was utilized to access the expression of these genes in the PE and normal blood samples. RESULTS Six ferroptosis-related genes involved in PE were obtained by overlapping 3661 genes most associated with PE, 565 DEGs between PE and normal samples, and 259 known ferroptosis-related genes. Among these genes, patients with PE displaying lower expression levels of NOS2 and higher expression levels of PTGS2 had a higher ferroptosis potential index. The expression pattern of NOS2 was consistent in the GSE48424 and GSE98224 datasets. RT-qPCR data confirmed that NOS2 expression was more significantly elevated in patients with PE than in those with a normal pregnancy. CONCLUSIONS Our study explored the diagnostic value of ferroptosis-related genes in PE, and identified NOS2 as the key gene linking ferroptosis and PE, suggesting a new candidate biomarker for early PE diagnosis.
Collapse
Affiliation(s)
- Shuangming Cai
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Shan Huang
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Wenni Zhang
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Huanshun Xiao
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Danfeng Yu
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Xuan Zhong
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Pei Tao
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China
| | - Yiping Luo
- Medical Intensive Care Unit, Guangdong Women and Children Hospital, Guangzhou, Guangdong, China.
| |
Collapse
|
9
|
Fothergill DM, Gertner JW. Exhaled Nitric Oxide and Pulmonary Oxygen Toxicity Susceptibility. Metabolites 2023; 13:930. [PMID: 37623874 PMCID: PMC10456729 DOI: 10.3390/metabo13080930] [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: 06/23/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/26/2023] Open
Abstract
Individual susceptibility to pulmonary oxygen toxicity (PO2tox) is highly variable and currently lacks a reliable biomarker for predicting pulmonary hyperoxic stress. As nitric oxide (NO) is involved in many respiratory system processes and functions, we aimed to determine if expired nitric oxide (FENO) levels can provide an indication of PO2tox susceptibility in humans. Eight U.S. Navy-trained divers volunteered as subjects. The hyperoxic exposures consisted of six- and eight-hour hyperbaric chamber dives conducted on consecutive days in which subjects breathed 100% oxygen at 202.65 kPa. Subjects' individual variability in pulmonary function and FENO was measured twice daily over five days and compared with their post-dive values to assess susceptibility to PO2tox. Only subjects who showed no decrements in pulmonary function following the six-hour exposure conducted the eight-hour dive. FENO decreased by 55% immediately following the six-hour oxygen exposure (n = 8, p < 0.0001) and by 63% following the eight-hour exposure (n = 4, p < 0.0001). Four subjects showed significant decreases in pulmonary function immediately following the six-hour exposure. These subjects had the lowest baseline FENO, had the lowest post-dive FENO, and had clinical symptoms of PO2tox. Individuals with low FENO were the first to develop PO2tox symptoms and deficits in pulmonary function from the hyperoxic exposures. These data suggest that endogenous levels of NO in the lungs may protect against the development of PO2tox.
Collapse
|
10
|
Sherratt SCR, Libby P, Dawoud H, Bhatt DL, Malinski T, Mason RP. Eicosapentaenoic acid (EPA) reduces pulmonary endothelial dysfunction and inflammation due to changes in protein expression during exposure to particulate matter air pollution. Biomed Pharmacother 2023; 162:114629. [PMID: 37027984 DOI: 10.1016/j.biopha.2023.114629] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/09/2023] Open
Abstract
AIMS Inhalation of air pollution small particle matter (PM) is a leading cause of cardiovascular (CV) disease. Exposure to PMs causes endothelial cell (EC) dysfunction as evidenced by nitric oxide (NO) synthase uncoupling, vasoconstriction and inflammation. Eicosapentaenoic acid (EPA) has been shown to mitigate PM-induced adverse cardiac changes in patients receiving omega-3 fatty acid supplementation. We set out to determine the pro-inflammatory effects of multiple PMs (urban and fine) on pulmonary EC NO bioavailability and protein expression, and whether EPA restores EC function under these conditions. METHODS AND RESULTS We pretreated pulmonary ECs with EPA and then exposed them to urban or fine air pollution PMs. LC/MS-based proteomic analysis to assess relative expression levels. Expression of adhesion molecules was measured by immunochemistry. The ratio of NO to peroxynitrite (ONOO-) release, an indication of eNOS coupling, was measured using porphyrinic nanosensors following calcium stimulation. Urban/fine PMs also modulated 9/12 and 13/36 proteins, respectively, linked to platelet and neutrophil degranulation pathways and caused > 50% (p < 0.001) decrease in the stimulated NO/ONOO- release ratio. EPA treatment altered expression of proteins involved in these inflammatory pathways, including a decrease in peroxiredoxin-5 and an increase in superoxide dismutase-1. EPA also increased expression of heme oxygenase-1 (HMOX1), a cytoprotective protein, by 2.1-fold (p = 0.024). EPA reduced elevations in sICAM-1 levels by 22% (p < 0.01) and improved the NO/ONOO- release ratio by > 35% (p < 0.05). CONCLUSION These cellular changes may contribute to anti-inflammatory, cytoprotective and lipid changes associated with EPA treatment during air pollution exposure.
Collapse
Affiliation(s)
- Samuel C R Sherratt
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, NH, USA; Elucida Research LLC, Beverly, MA, USA
| | - Peter Libby
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Hazem Dawoud
- Nanomedical Research Laboratory, Ohio University, Athens, OH, USA
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Health System, New York, NY, USA
| | - Tadeusz Malinski
- Nanomedical Research Laboratory, Ohio University, Athens, OH, USA.
| | - R Preston Mason
- Elucida Research LLC, Beverly, MA, USA; Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
González-Candia A, Candia AA, Arias PV, Paz AA, Herrera EA, Castillo RL. Chronic intermittent hypobaric hypoxia induces cardiovascular dysfunction in a high-altitude working shift model. Life Sci 2023:121800. [PMID: 37245841 DOI: 10.1016/j.lfs.2023.121800] [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: 02/03/2023] [Revised: 05/17/2023] [Accepted: 05/20/2023] [Indexed: 05/30/2023]
Abstract
AIMS Chronic intermittent hypobaric hypoxia (CIHH) exposure due to shift work occurs mainly in 4 × 4 or 7 × 7 days shifts in mining, astronomy, and customs activities, among other institutions. However, the long-lasting effects of CIHH on cardiovascular structure and function are not well characterized. We aimed to investigate the effects of CIHH on the cardiac and vascular response of adult rats simulating high-altitude (4600 m) x low-altitude (760 m) working shifts. MAIN METHODS We analyzed in vivo cardiac function through echocardiography, ex vivo vascular reactivity by wire myography, and in vitro cardiac morphology by histology and protein expression and immunolocalization by molecular biology and immunohistochemistry techniques in 12 rats, 6 exposed to CIHH in the hypoxic chamber, and respective normobaric normoxic controls (n = 6). KEY FINDINGS CIHH induced cardiac dysfunction with left and right ventricle remodeling, associated with an increased collagen content in the right ventricle. In addition, CIHH increased HIF-1α levels in both ventricles. These changes are associated with decreased antioxidant capacity in cardiac tissue. Conversely, CIHH decreased contractile capacity with a marked decreased in nitric oxide-dependent vasodilation in both, carotid and femoral arteries. SIGNIFICANCE These data suggest that CIHH induces cardiac and vascular dysfunction by ventricular remodeling and impaired vascular vasodilator function. Our findings highlight the impact of CIHH in cardiovascular function and the importance of a periodic cardiovascular evaluation in high-altitude workers.
Collapse
Affiliation(s)
| | - Alejandro A Candia
- Institute of Health Sciences, University of O'Higgins, Rancagua, Chile; Department for the Woman and Newborn Health Promotion, Universidad de Chile, Chile
| | - Pamela V Arias
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Adolfo A Paz
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Emilio A Herrera
- Laboratory of Vascular Function & Reactivity, Pathophysiology Program, ICBM, Faculty of Medicine, Universidad de Chile, Santiago, Chile; International Center for Andean Studies (INCAS), University of Chile, Putre, Chile.
| | - Rodrigo L Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago, Chile; Unidad de Paciente Crítico, Hospital del Salvador, Santiago, Chile.
| |
Collapse
|
12
|
Chen Q, Guo J, Qiu T, Zhou J. Mechanism of ASK1 involvement in liver diseases and related potential therapeutic targets: A critical pathway molecule worth investigating. J Gastroenterol Hepatol 2023; 38:378-385. [PMID: 36533997 DOI: 10.1111/jgh.16087] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/25/2022] [Accepted: 12/10/2022] [Indexed: 12/30/2022]
Abstract
Since the discovery of apoptosis signal-regulated kinase 1 (ASK1), the signal transduction mechanism and pathophysiological process involved in its regulation have been continuously revealed. Many previous studies have identified that ASK1 is involved and plays a critical role in the development of diseases affecting the nervous, cardiac, renal, and other systems. As a mitogen-activated protein kinase (MAPK) kinase kinase, ASK1 mediates apoptosis, necrosis, inflammation, and other pathological processes by activating its downstream c-Jun N-terminal kinase (JNK)/p38 MAPK. Owing to the important role of ASK1, an increasing number of studies in recent years have focused on its status in liver-related diseases. In this paper, we review the mechanisms and targets of ASK1 in liver-related diseases to emphasize its important role in the development of liver disease.
Collapse
Affiliation(s)
- Qi Chen
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| | - Jiayu Guo
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| | - Tao Qiu
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| | - Jiangqiao Zhou
- Department of Organ Transplantation, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, Hubei Province, China
| |
Collapse
|
13
|
Age-Related Changes in Skeletal Muscle Oxygen Utilization. J Funct Morphol Kinesiol 2022; 7:jfmk7040087. [PMID: 36278748 PMCID: PMC9590092 DOI: 10.3390/jfmk7040087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
The cardiovascular and skeletal muscle systems are intrinsically interconnected, sharing the goal of delivering oxygen to metabolically active tissue. Deficiencies within those systems that affect oxygen delivery to working tissues are a hallmark of advancing age. Oxygen delivery and utilization are reflected as muscle oxygen saturation (SmO2) and are assessed using near-infrared resonance spectroscopy (NIRS). SmO2 has been observed to be reduced by ~38% at rest, ~24% during submaximal exercise, and ~59% during maximal exercise with aging (>65 y). Furthermore, aging prolongs restoration of SmO2 back to baseline by >50% after intense exercise. Regulatory factors that contribute to reduced SmO2 with age include blood flow, capillarization, endothelial cells, nitric oxide, and mitochondrial function. These mechanisms are governed by reactive oxygen species (ROS) at the cellular level. However, mishandling of ROS with age ultimately leads to alterations in structure and function of the regulatory factors tasked with maintaining SmO2. The purpose of this review is to provide an update on the current state of the literature regarding age-related effects in SmO2. Furthermore, we attempt to bridge the gap between SmO2 and associated underlying mechanisms affected by aging.
Collapse
|
14
|
Doman AJ, Tommasi S, Perkins MV, McKinnon RA, Mangoni AA, Nair PC. Chemical similarities and differences among inhibitors of nitric oxide synthase, arginase and dimethylarginine dimethylaminohydrolase-1: implications for the design of novel enzyme inhibitors modulating the nitric oxide pathway. Bioorg Med Chem 2022; 72:116970. [DOI: 10.1016/j.bmc.2022.116970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 08/01/2022] [Accepted: 08/18/2022] [Indexed: 11/02/2022]
|
15
|
John RV, Devasia T, N M, Lukose J, Chidangil S. Micro-Raman spectroscopy study of blood samples from myocardial infarction patients. Lasers Med Sci 2022; 37:3451-3460. [PMID: 35821543 DOI: 10.1007/s10103-022-03604-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 06/28/2022] [Indexed: 12/15/2022]
Abstract
Acute myocardial infarction (MI) is found to be a major causative factor for global mortality and morbidity. This situation demands necessity of developing efficient and rapid diagnostic tools to detect acute MI. Raman spectroscopy is a non-destructive optical diagnostic technique, which has high potential in probing biochemical changes in clinical samples during initiation and progress of diseases. In this work, blood was taken as the sample to examine inflammation in acute MI patients using Raman spectroscopy. Ratio of Raman peak intensities that corresponds to phenylalanine (1000 cm-1) and tyrosine (825 cm-1) can facilitate indirect information about tetrahydrobiopterin (BH4) availability, which can indicate inflammatory status in patients. This ratio obtained was higher for MI patients in comparison with control subjects. The decrease in phenylalanine and tyrosine ratio (Phe-Tyr ratio) is attributed to the prognosis of standard of care (medications like antiplatelets including aspirin, statin and revascularisation) leading to inflammation reduction. Phe-Tyr ratio estimated from the Raman spectra of blood can be exploited as a reliable method to probe inflammation due to MI. The method is highly objective, require only microliters of sample and minimal sample preparation, signifying its clinical utility.
Collapse
Affiliation(s)
- Reena V John
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Tom Devasia
- Department of Cardiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Mithun N
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jijo Lukose
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Santhosh Chidangil
- Centre of Excellence for Biophotonics, Department of Atomic and Molecular Physics, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
16
|
Bouly M, Bourguignon MP, Roesch S, Rigouin P, Gosgnach W, Bossard E, Royere E, Diguet N, Sansilvestri-Morel P, Bonnin A, Xuereb L, Berson P, Komajda M, Bernhardt P, Tyl B. Aging increases circulating BH 2 without modifying BH 4 levels and impairs peripheral vascular function in healthy adults. Transl Res 2021; 238:36-48. [PMID: 34332154 DOI: 10.1016/j.trsl.2021.07.004] [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: 04/09/2021] [Revised: 06/23/2021] [Accepted: 07/23/2021] [Indexed: 11/17/2022]
Abstract
Little is known about the mechanisms of aging on vascular beds and its relationship with tetra and di-hydrobiopterin (BH4 and BH2) levels. This observational clinical study analyzed the impact of aging on plasma and platelet biopterins, cutaneous blood flow (CBF), and coronary flow reserve (CFR) in healthy adults. The study enrolled healthy adults in 3 age groups: 18-30, 50-59, and 60-70 years (n = 25/group). Biopterins were assessed by LC-MS/MS using newly defined pre-analytical conditions limiting BH4 oxidation and improving long-term stability. CBF was measured by Laser Speckle Contrast Imaging coupled with acetylcholine-iontophoresis and CFR by adenosine stress cardiac magnetic resonance. In healthy adults, aging (60-70 years vs 18-30 years) significantly increased platelet BH2 (+75%, P = 0.033) and BH2 + BH4 (+31%, P = 0.033), and to a lesser extent plasma BH2 (+29%, P = 0.009) without affecting BH4 and BH4/BH2. Simultaneously, CBF was decreased (-23%, P = 0.004) but not CFR, CBF being inversely correlated with platelet BH2 (r = -0.42, P = 0.001) and BH2 + BH4 (r = -0.41, P = 0.002). The proportion of adults with abnormal platelet BH2 increased with age (+28% in 60-70y). These abnormal BH2 levels were significantly associated with reduced CBF and CFR (-16%, P = 0.03 and -26%, P = 0.02). In conclusion, our study showed that age-related peripheral endothelial dysfunction was associated with an increase in circulating BH2 without decreasing BH4, the effect being more marked in platelets, the most relevant blood compartment to assess biopterin bioavailability. Peripheral but not coronary vascular function is progressively impaired with aging in healthy adults. All these findings support biopterins as therapeutic targets to improve vascular function.
Collapse
Affiliation(s)
- Muriel Bouly
- Cardiovascular & Metabolic Disease Centre for Therapeutic Innovation, Institut de Recherches Internationales Servier, Suresnes, France
| | - Marie-Pierre Bourguignon
- Cardiovascular & Metabolic Disease Centre for Therapeutic Innovation, Institut de Recherches Servier, Suresnes, France
| | - Susanne Roesch
- Centre of Excellence Clinical Operations, Institut de Recherches Internationales Servier, Suresnes, France
| | - Pascal Rigouin
- Biostatistics Department, Keyrus Life Science, Nantes, France
| | - Willy Gosgnach
- Centre of Excellence Biotechnology, Institut de Recherches Servier, Croissy, France
| | | | - Emilie Royere
- Cardiovascular & Metabolic Disease Centre for Therapeutic Innovation, Institut de Recherches Servier, Suresnes, France
| | - Nicolas Diguet
- Cardiovascular & Metabolic Disease Centre for Therapeutic Innovation, Institut de Recherches Servier, Suresnes, France
| | - Patricia Sansilvestri-Morel
- Cardiovascular & Metabolic Disease Centre for Therapeutic Innovation, Institut de Recherches Servier, Suresnes, France
| | - Ariane Bonnin
- Drug Safety & Pharmacokinetics Centre of Excellence, Biologie Servier, Gidy, France
| | - Laura Xuereb
- Centre of Excellence Methodology and Valorisation of Data, Institut de Recherches Internationales Servier, Suresnes, France
| | - Pascal Berson
- Cardiovascular & Metabolic Disease Centre for Therapeutic Innovation, Institut de Recherches Servier, Suresnes, France
| | - Michel Komajda
- Department of Cardiology, Hospital Saint Joseph, France/Paris Sorbonne Université France, Paris, France
| | | | - Benoit Tyl
- Cardiovascular & Metabolic Disease Centre for Therapeutic Innovation, Institut de Recherches Internationales Servier, Suresnes, France
| |
Collapse
|
17
|
Yao F, Abdel-Rahman AA. Tetrahydrobiopterin paradoxically mediates cardiac oxidative stress and mitigates ethanol-evoked cardiac dysfunction in conscious female rats. Eur J Pharmacol 2021; 909:174406. [PMID: 34364878 PMCID: PMC8434968 DOI: 10.1016/j.ejphar.2021.174406] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 02/04/2023]
Abstract
Oxidation of tetrahydrobiopterin (BH4), a cofactor of nitric oxide synthase (NOS), by reactive oxidative species (ROS), leads to NOS uncoupling and superoxide production instead of NO. Further, oxidative stress plays a major role in ethanol-evoked cardiac dysfunction in proestrus female rats, and acute ethanol administration reduces brain BH4 level. Therefore, we discerned the unknown role of BH4 in ethanol-evoked cardiac dysfunction by pharmacologically increasing BH4 levels or inhibiting its effect in proestrus female rats. Acute ethanol (1.5 g/kg, i.v, 30 min) caused myocardial dysfunction (lowered dP/dtmax and LVDP) and hypotension, along with increases in myocardial: (i) levels of NO, ROS and malondialdehyde (MDA), (ii) activities of catalase, ALDH2 and NADPH oxidase (Nox), and (iii) phosphorylation of eNOS, nNOS. Further, ethanol suppressed myocardial arginase and superoxide dismutase (SOD) activities and enhanced eNOS uncoupling. While ethanol had no effect on cardiac BH4 levels, BH4 (19 mg/kg, i.v) supplementation paradoxically caused cardiac oxidative stress, but mitigated the cardiac dysfunction/hypotension and most of the adverse molecular responses caused by ethanol. Equally important, the BH4 inhibitor DAHP (1 g/kg, i.p) exacerbated the adverse molecular and cardiovascular effects caused by ethanol. Our pharmacological studies support a protective role for the NOS co-factor BH4 against ethanol-evoked cardiac dysfunction and hypotension in female rats.
Collapse
Affiliation(s)
- Fanrong Yao
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA
| | - Abdel A Abdel-Rahman
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC, 27834, USA.
| |
Collapse
|
18
|
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
|
19
|
Ow CPC, Trask-Marino A, Betrie AH, Evans RG, May CN, Lankadeva YR. Targeting Oxidative Stress in Septic Acute Kidney Injury: From Theory to Practice. J Clin Med 2021; 10:jcm10173798. [PMID: 34501245 PMCID: PMC8432047 DOI: 10.3390/jcm10173798] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/17/2021] [Accepted: 08/23/2021] [Indexed: 12/17/2022] Open
Abstract
Sepsis is the leading cause of acute kidney injury (AKI) and leads to increased morbidity and mortality in intensive care units. Current treatments for septic AKI are largely supportive and are not targeted towards its pathophysiology. Sepsis is commonly characterized by systemic inflammation and increased production of reactive oxygen species (ROS), particularly superoxide. Concomitantly released nitric oxide (NO) then reacts with superoxide, leading to the formation of reactive nitrogen species (RNS), predominantly peroxynitrite. Sepsis-induced ROS and RNS can reduce the bioavailability of NO, mediating renal microcirculatory abnormalities, localized tissue hypoxia and mitochondrial dysfunction, thereby initiating a propagating cycle of cellular injury culminating in AKI. In this review, we discuss the various sources of ROS during sepsis and their pathophysiological interactions with the immune system, microcirculation and mitochondria that can lead to the development of AKI. We also discuss the therapeutic utility of N-acetylcysteine and potential reasons for its efficacy in animal models of sepsis, and its inefficacy in ameliorating oxidative stress-induced organ dysfunction in human sepsis. Finally, we review the pre-clinical studies examining the antioxidant and pleiotropic actions of vitamin C that may be of benefit for mitigating septic AKI, including future implications for clinical sepsis.
Collapse
Affiliation(s)
- Connie P. C. Ow
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Cardiac Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka 564-8565, Japan
| | - Anton Trask-Marino
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
| | - Ashenafi H. Betrie
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia
| | - Roger G. Evans
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Cardiovascular Disease Program, Biomedicine Discovery Institute and Department of Physiology, Monash University, Melbourne, VIC 3800, Australia
| | - Clive N. May
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Yugeesh R. Lankadeva
- Preclinical Critical Care Unit, Florey Institute of Neuroscience and Mental Health, Melbourne, VIC 3052, Australia; (C.P.C.O.); (A.T.-M.); (A.H.B.); (R.G.E.); (C.N.M.)
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC 3052, Australia
- Correspondence: ; Tel.: +61-3-8344-0417; Fax: +61-3-9035-3107
| |
Collapse
|
20
|
Zhong GC, Zhao ZB, Cheng Y, Wang YB, Qiu C, Mao LH, Hu JJ, Cai D, Liu Y, Gong JP, Li SW. Epigenetic silencing of GCH1promotes hepatocellular carcinoma growth by activating superoxide anion-mediated ASK1/p38 signaling via inhibiting tetrahydrobiopterin de novo biosynthesis. Free Radic Biol Med 2021; 168:81-94. [PMID: 33781891 DOI: 10.1016/j.freeradbiomed.2021.03.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/21/2022]
Abstract
Metabolic reprogramming is a hallmark of cancer, including hepatocellular carcinoma (HCC). However, its role in HCC remains to be elucidated. Herein, we identified GTP cyclohydrolase 1 (GCH1), the first rate-limiting enzyme in tetrahydrobiopterin (BH4) de novo biosynthesis, as a novel metabolic regulator of HCC. GCH1 was frequently down-regulated in HCC tissues and cell lines by promoter methylation. Low GCH1 expression was associated with larger tumor size, increased tumor number, and worse prognosis in two independent cohorts of HCC patients. Functionally, GCH1 silencing promoted HCC growth in vitro and in vivo, while GCH1 overexpression exerted an opposite effect. The metabolite BH4 inhibited HCC growth in vitro and in vivo. GCH1 silencing exerted its growth-promoting effect through directly inhibiting BH4 de novo biosynthesis. Mechanistically, GCH1 silencing activated ASK1/p38 signaling; pharmacological or genetic inhibition of ASK1 or p38 abolished GCH1 silencing-induced growth-promoting effect. Further mechanistic studies found that GCH1 silencing-induced BH4 reduction resulted in an increase of intracellular superoxide anion levels in a dose-dependent manner, which mediated the activation of ASK1/p38 signaling. Collectively, our study reveals that epigenetic silencing of GCH1 promotes HCC growth by activating superoxide anion-mediated ASK1/p38 signaling via inhibiting BH4 de novo biosynthesis, suggesting that targeting GCH1/BH4 pathway may be a promising therapeutic strategy to combat HCC.
Collapse
Affiliation(s)
- Guo-Chao Zhong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhi-Bo Zhao
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yao Cheng
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yun-Bing Wang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chan Qiu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Hong Mao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie-Jun Hu
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Cai
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yan Liu
- Department of Gastroenterology, The Fifth People's Hospital of Chengdu, Chengdu, China
| | - Jian-Ping Gong
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Sheng-Wei Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
21
|
Galler KM, Weber M, Korkmaz Y, Widbiller M, Feuerer M. Inflammatory Response Mechanisms of the Dentine-Pulp Complex and the Periapical Tissues. Int J Mol Sci 2021; 22:ijms22031480. [PMID: 33540711 PMCID: PMC7867227 DOI: 10.3390/ijms22031480] [Citation(s) in RCA: 196] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/25/2021] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
The macroscopic and microscopic anatomy of the oral cavity is complex and unique in the human body. Soft-tissue structures are in close interaction with mineralized bone, but also dentine, cementum and enamel of our teeth. These are exposed to intense mechanical and chemical stress as well as to dense microbiologic colonization. Teeth are susceptible to damage, most commonly to caries, where microorganisms from the oral cavity degrade the mineralized tissues of enamel and dentine and invade the soft connective tissue at the core, the dental pulp. However, the pulp is well-equipped to sense and fend off bacteria and their products and mounts various and intricate defense mechanisms. The front rank is formed by a layer of odontoblasts, which line the pulp chamber towards the dentine. These highly specialized cells not only form mineralized tissue but exert important functions as barrier cells. They recognize pathogens early in the process, secrete antibacterial compounds and neutralize bacterial toxins, initiate the immune response and alert other key players of the host defense. As bacteria get closer to the pulp, additional cell types of the pulp, including fibroblasts, stem and immune cells, but also vascular and neuronal networks, contribute with a variety of distinct defense mechanisms, and inflammatory response mechanisms are critical for tissue homeostasis. Still, without therapeutic intervention, a deep carious lesion may lead to tissue necrosis, which allows bacteria to populate the root canal system and invade the periradicular bone via the apical foramen at the root tip. The periodontal tissues and alveolar bone react to the insult with an inflammatory response, most commonly by the formation of an apical granuloma. Healing can occur after pathogen removal, which is achieved by disinfection and obturation of the pulp space by root canal treatment. This review highlights the various mechanisms of pathogen recognition and defense of dental pulp cells and periradicular tissues, explains the different cell types involved in the immune response and discusses the mechanisms of healing and repair, pointing out the close links between inflammation and regeneration as well as between inflammation and potential malignant transformation.
Collapse
Affiliation(s)
- Kerstin M. Galler
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, 93093 Regensburg, Germany;
- Correspondence:
| | - Manuel Weber
- Department of Oral and Maxillofacial Surgery, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Yüksel Korkmaz
- Department of Periodontology and Operative Dentistry, University of Mainz, 55131 Mainz, Germany;
| | - Matthias Widbiller
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, 93093 Regensburg, Germany;
| | - Markus Feuerer
- Department for Immunology, University Hospital Regensburg, 93053 Regensburg, Germany;
- Regensburg Center for Interventional Immunology (RCI), University Hospital Regensburg, 93053 Regensburg, Germany
| |
Collapse
|
22
|
Watanabe T. Neopterin derivatives - a novel therapeutic target rather than biomarker for atherosclerosis and related diseases. VASA 2020; 50:165-173. [PMID: 32924886 DOI: 10.1024/0301-1526/a000903] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This review provides an updated overview of the emerging roles of neopterin derivatives in atherosclerosis. Neopterin, a metabolite of guanosine triphosphate, is produced by interferon-γ-activated macrophages and is expressed at high levels in atheromatous plaques within the human carotid and coronary arteries as well as in the aorta. Plasma concentrations of neopterin are higher in patients with carotid, cerebral, and coronary artery diseases as well as aortic aneurysm. The concentration of neopterin is positively correlated with the severity of coronary artery disease. However, a prospective cohort study showed that neopterin contributes to protection against plaque formation in carotid arteries in patients with atherosclerosis. Moreover, using both in vitro and in vivo experiments, a recent study has shown the atheroprotective effects of neopterin. Neopterin suppresses the expression of monocyte chemotactic protein-1, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1 in endothelial cells, and thereby suppresses the adhesion of monocytes to endothelial cells. It also suppresses the inflammatory phenotype of monocyte-derived macrophages. In addition, neopterin suppresses oxidized low-density lipoprotein-induced foam cell formation in macrophages and the migration and proliferation of vascular smooth muscle cells. Neopterin injection into apolipoprotein E-deficient (Apoe-/-) mice suppresses the development of atherosclerotic lesions. A neopterin derivative tetrahydroneopterin (BH4), also known as a cofactor for nitric oxide (NO) synthases, suppresses atherosclerosis and vascular injury-induced neointimal hyperplasia in Apoe-/- mice. BH4 administration improves endothelial dysfunction in patients with coronary artery disease. These findings suggest that neopterin production may increase to counteract the progression of atherosclerosis, as neopterin contributes to atheroprotection. Otherwise, the increased neopterin levels in atherosclerosis may reflect a compensatory mechanism associated with inducible NO synthase upregulation in macrophages to supply BH4 for high output NO production caused by decreased endothelial NO synthase in atherosclerosis. Therefore, neopterin derivatives are a novel therapeutic target for atherosclerosis and related diseases.
Collapse
Affiliation(s)
- Takuya Watanabe
- Department of Internal Medicine, Ushioda General Hospital/Clinic, Yokohama, Japan
| |
Collapse
|
23
|
Palve V, Liao Y, Remsing Rix LL, Rix U. Turning liabilities into opportunities: Off-target based drug repurposing in cancer. Semin Cancer Biol 2020; 68:209-229. [PMID: 32044472 DOI: 10.1016/j.semcancer.2020.02.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022]
Abstract
Targeted drugs and precision medicine have transformed the landscape of cancer therapy and significantly improved patient outcomes in many cases. However, as therapies are becoming more and more tailored to smaller patient populations and acquired resistance is limiting the duration of clinical responses, there is an ever increasing demand for new drugs, which is not easily met considering steadily rising drug attrition rates and development costs. Considering these challenges drug repurposing is an attractive complementary approach to traditional drug discovery that can satisfy some of these needs. This is facilitated by the fact that most targeted drugs, despite their implicit connotation, are not singularly specific, but rather display a wide spectrum of target selectivity. Importantly, some of the unintended drug "off-targets" are known anticancer targets in their own right. Others are becoming recognized as such in the process of elucidating off-target mechanisms that in fact are responsible for a drug's anticancer activity, thereby revealing potentially new cancer vulnerabilities. Harnessing such beneficial off-target effects can therefore lead to novel and promising precision medicine approaches. Here, we will discuss experimental and computational methods that are employed to specifically develop single target and network-based off-target repurposing strategies, for instance with drug combinations or polypharmacology drugs. By illustrating concrete examples that have led to clinical translation we will furthermore examine the various scientific and non-scientific factors that cumulatively determine the success of these efforts and thus can inform the future development of new and potentially lifesaving off-target based drug repurposing strategies for cancers that constitute important unmet medical needs.
Collapse
Affiliation(s)
- Vinayak Palve
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Yi Liao
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
24
|
Nitric oxide: Is it the culprit for the continued expansion of keloids? Eur J Pharmacol 2019; 854:282-288. [PMID: 31034822 DOI: 10.1016/j.ejphar.2019.04.040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/19/2019] [Accepted: 04/25/2019] [Indexed: 11/22/2022]
Abstract
Keloids are characterized by excessive proliferation of fibroblasts and invasion of surrounding healthy skin. High levels of Nitric Oxide (NO) are thought to be the crucial factor within the micro-environment in promoting keloid formation. However, the effects and mechanisms of NO on the proliferation of Keloid Fibroblasts (KDFs) remain unclear. In this study, we investigated the effect of NO on KDFs proliferation by Sodium Nitroprusside (SNP), an NO donor. Our results show that SNP significantly enhanced KDFs proliferation. Moreover, with prolonged treatment with SNP after cell confluence, the growth of KDFs escape contact inhibition and experience significant pile up growth. Furthermore, PTIO, an NO scavenger, attenuated SNP-enhanced cell proliferation effectively. The mechanism involved in SNP-induced KDFs proliferation was soluble Guanylyl Cyclase (sGC) and cGMP independent. ODQ, a specific sGC inhibitor, failed to suppress SNP-enhanced KDFs proliferation. 8-Bromo-c GMP, a cell-permeable cGMP analogue, could not stimulate KDFs proliferation. Erk and Akt provide important signaling for cell growth. U0126 and LY294002, inhibitors of Erk and Akt respectively, block SNP-enhanced KDFs proliferation effectively. As expected, a Western blot showed that SNP up-regulated the phosphorylation levels of Erk and Akt. Moreover, it decreased the expression of p27, a cell cycle inhibitor. Our results reveal that SNP induced KDFs proliferation and loss contact inhibition led to pile up growth via activation of the Erk and Akt pathways, as well as a decreased expression of p27. Thus, we speculate that the pathological feature of continuous expansion in keloids is caused by NO-induced KDFs sustained growth.
Collapse
|
25
|
Xie L, Hu D, Qin H, Zhang W, Zhang S, Feng Y, Yao H, Xiao Y, Yao K, Huang X. In vivo gum arabic-coated tetrahydrobiopterin protects against myocardial ischemia reperfusion injury by preserving eNOS coupling. Life Sci 2019; 219:294-302. [PMID: 30668954 DOI: 10.1016/j.lfs.2019.01.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 01/10/2019] [Accepted: 01/17/2019] [Indexed: 12/23/2022]
Abstract
AIMS Exogenous tetrahydrobiopterin (BH4), an indispensable cofactor of endothelial nitric oxide synthase (eNOS), supplementation has been proved to be of advantage to improve cardiovascular function. Nevertheless, due to its highly redox-sensitive and easy to be oxidized, there is an urgent need to develop an appropriate BH4 formulation for clinical therapy. Gum Arabic (GA) has been considered as an alternative biopolymer for the stabilization and coating of drugs. The effects of GA on protecting BH4 from being oxidized were investigated in a rat model of myocardial ischemia-reperfusion (I/R). MAIN METHODS Rats were subjected to 60-min of in vivo left coronary artery occlusion and varying periods of reperfusion with or without pre-ischemic GA-coated BH4 supplementation (10 mg/kg, oral). Myocardial infarction, fibrotic area and left ventricle ejection fraction were correlated with cardiac BH4 content, eNOS protein, NOS enzyme activity, and ROS/NO generation. KEY FINDINGS Pretreatment of rats with GA-coated 6R-BH4, 24 h before myocardial ischemia, resulted in smaller myocardial infarction, improved left ventricular function and inhibited fibrosis, correlated with maintained high levels of cardiac BH4 content, preserved eNOS activation and dimerization, and decreased ROS generation. However in uncoated group, 6R-BH4 treatment did not reduce acute and chronic myocardial I/R injury compared with control I/R rats, which was closely related with the marked loss of myocardial BH4 levels during I/R. SIGNIFICANCE These findings provide evidence that in vivo pre-ischemic oral GA-coated BH4 administration preserves eNOS function secondary to maintaining cardiac BH4 content, and confers cardioprotection after I/R.
Collapse
Affiliation(s)
- Lin Xie
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, China.
| | - Dan Hu
- Department of Neurology, Renmin Hospital of Wuhan University, China
| | - Huan Qin
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Wenliang Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Shiyao Zhang
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Yuan Feng
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Haozhe Yao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Ying Xiao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China
| | - Kai Yao
- Institute of Biology and Medicine, College of Life and Health Sciences, Wuhan University of Science and Technology, Hubei, China.
| | - Xia Huang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Key Laboratory of Organ Transplantation, Ministry of Education, China; NHC Key Laboratory of Organ Transplantation, China; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, China
| |
Collapse
|
26
|
Imrie H, Williams DJL. Stimulation of bovine monocyte-derived macrophages with lipopolysaccharide, interferon-ɣ, Interleukin-4 or Interleukin-13 does not induce detectable changes in nitric oxide or arginase activity. BMC Vet Res 2019; 15:45. [PMID: 30704453 PMCID: PMC6357487 DOI: 10.1186/s12917-019-1785-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 01/16/2019] [Indexed: 02/27/2023] Open
Abstract
BACKGROUND Bacterial lipopolysaccharide and interferon-γ stimulation of rodent macrophages in vitro induces up-regulation of inducible nitric oxide synthase, whereas interleukin-4 stimulation results in increased activity of arginase-1. Thus different stimulants result in differing macrophage phenotypes, appropriate for responses to a range of pathogens. The current study was conducted in order to determine whether bovine macrophages derived from monocytes and spleen respond similarly. RESULTS Lipopolysaccharide and interferon-γ did not induce detectable increases in nitric oxide production by bovine monocyte-derived or splenic macrophages in vitro. Similarly, interleukin-4 and interleukin-13 did not affect arginase activity. However, changes in transcription of genes coding for these products were detected. CONCLUSION Differences between macrophage activation patterns exist between cattle and other species and these differences may occur during the post-transcription phase.
Collapse
Affiliation(s)
- Heather Imrie
- Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF UK
- Animal, Rural and Environmental Sciences, Nottingham Trent University, Brackenhurst Lane, Southwell, NG25 0DT UK
| | - Diana J. L. Williams
- Institute of Infection and Global Health, University of Liverpool, 146 Brownlow Hill, Liverpool, L3 5RF UK
| |
Collapse
|
27
|
McNeill E, Stylianou E, Crabtree MJ, Harrington-Kandt R, Kolb AL, Diotallevi M, Hale AB, Bettencourt P, Tanner R, O'Shea MK, Matsumiya M, Lockstone H, Müller J, Fletcher HA, Greaves DR, McShane H, Channon KM. Regulation of mycobacterial infection by macrophage Gch1 and tetrahydrobiopterin. Nat Commun 2018; 9:5409. [PMID: 30573728 PMCID: PMC6302098 DOI: 10.1038/s41467-018-07714-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
Inducible nitric oxide synthase (iNOS) plays a crucial role in controlling growth of Mycobacterium tuberculosis (M.tb), presumably via nitric oxide (NO) mediated killing. Here we show that leukocyte-specific deficiency of NO production, through targeted loss of the iNOS cofactor tetrahydrobiopterin (BH4), results in enhanced control of M.tb infection; by contrast, loss of iNOS renders mice susceptible to M.tb. By comparing two complementary NO-deficient models, Nos2-/- mice and BH4 deficient Gch1fl/flTie2cre mice, we uncover NO-independent mechanisms of anti-mycobacterial immunity. In both murine and human leukocytes, decreased Gch1 expression correlates with enhanced cell-intrinsic control of mycobacterial infection in vitro. Gene expression analysis reveals that Gch1 deficient macrophages have altered inflammatory response, lysosomal function, cell survival and cellular metabolism, thereby enhancing the control of bacterial infection. Our data thus highlight the importance of the NO-independent functions of Nos2 and Gch1 in mycobacterial control.
Collapse
Affiliation(s)
- Eileen McNeill
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.
| | | | - Mark J Crabtree
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | | | - Anna-Lena Kolb
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Marina Diotallevi
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Ashley B Hale
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | | | - Rachel Tanner
- Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | | | | | - Helen Lockstone
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK
| | - Julius Müller
- Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Helen A Fletcher
- Department of Immunology and Infection, London School of Hygiene and Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - David R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK
| | - Helen McShane
- Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Keith M Channon
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, OX3 7BN, UK.
| |
Collapse
|
28
|
Rivera JC, Noueihed B, Madaan A, Lahaie I, Pan J, Belik J, Chemtob S. Tetrahydrobiopterin (BH4) deficiency is associated with augmented inflammation and microvascular degeneration in the retina. J Neuroinflammation 2017; 14:181. [PMID: 28874201 PMCID: PMC5586016 DOI: 10.1186/s12974-017-0955-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 08/28/2017] [Indexed: 02/07/2023] Open
Abstract
Background Tetrahydrobiopterin (BH4) is an essential cofactor in multiple metabolic processes and plays an essential role in maintaining the inflammatory and neurovascular homeostasis. In this study, we have investigated the deleterious effects of BH4 deficiency on retinal vasculature during development. Methods hph-1 mice, which display deficiency in BH4 synthesis, were used to characterize the inflammatory effects and the integrity of retinal microvasculature. BH4 levels in retinas from hph-1 and wild type (WT) mice were measured by LC-MS/MS. Retinal microvascular area and microglial cells number were quantified in hph-1 and WT mice at different ages. Retinal expression of pro-inflammatory, anti-angiogenic, and neuronal-derived factors was analyzed by qPCR. BH4 supplementation was evaluated in vitro, ex-vivo, and in vivo models. Results Our findings demonstrated that BH4 levels in the retina from hph-1 mice were significantly lower by ~ 90% at all ages analyzed compared to WT mice. Juvenile hph-1 mice showed iris atrophy, persistent fetal vasculature, significant increase in the number of microglial cells (p < 0.01), as well as a marked degeneration of the retinal microvasculature. Retinal microvascular alterations in juvenile hph-1 mice were associated with a decreased expression in Norrin (0.2-fold) and its receptor Frizzled-4 (FZD4; 0.51-fold), as well as with an augmented expression of pro-inflammatory factors such as IL-6 (3.2-fold), NRLP-3 (4.4-fold), IL-1β (8.6-fold), and the anti-angiogenic factor thrombospondin-1 (TSP-1; 17.5-fold). We found that TSP-1 derived from activated microglial cells is a factor responsible of inducing microvascular degeneration, but BH4 supplementation markedly prevented hyperoxia-induced microglial activation in vitro and microvascular injury in an ex-vivo model of microvascular angiogenesis and an in vivo model of oxygen-induced retinopathy (OIR). Conclusion Our findings reveal that BH4 is a key cofactor in regulating the expression of inflammatory and anti-angiogenic factors that play an important function in the maintenance of retinal microvasculature. Electronic supplementary material The online version of this article (10.1186/s12974-017-0955-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- José Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, Université de Montréal, 5415 Blvd de l'Assomption, Montréal, Québec, H1T 2M4, Canada. .,Department of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada.
| | - Baraa Noueihed
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, Université de Montréal, 5415 Blvd de l'Assomption, Montréal, Québec, H1T 2M4, Canada
| | - Ankush Madaan
- Department of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada
| | - Isabelle Lahaie
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, Université de Montréal, 5415 Blvd de l'Assomption, Montréal, Québec, H1T 2M4, Canada
| | - Jingyi Pan
- Departments of Pediatrics and Physiology, The Hospital For Sick Children, University of Toronto, Toronto, Canada
| | - Jaques Belik
- Departments of Pediatrics and Physiology, The Hospital For Sick Children, University of Toronto, Toronto, Canada
| | - Sylvain Chemtob
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Center, Université de Montréal, 5415 Blvd de l'Assomption, Montréal, Québec, H1T 2M4, Canada. .,Department of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Center, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
29
|
Abstract
Endothelial cells are a constitutive part of the heart and vasculature and form a crucial link between the cardiovascular system and the immune system. Besides their commonly accepted roles in angiogenesis, hemostasis, and the regulation of vascular tone, they are an essential and active component of immune responses. Expression of a range of innate pattern recognition receptors allows them to respond to inflammatory stimulation, and they control immune cell recruitment and extravasation into target tissues throughout the body.In this chapter, I will therefore summarize classical endothelial cell properties and functions and their cross talk with the immune system as well as the operational immunological role of endothelial cells in facilitating immune responses.
Collapse
Affiliation(s)
- Caterina Sturtzel
- Innovative Cancer Models, Children's Cancer Research Institute, St. Anna Kinderkrebsforschung e.V, Vienna, Austria.
| |
Collapse
|
30
|
Tonin TD, Thiesen LC, de Oliveira Nunes ML, Broering MF, Donato MP, Goss MJ, Petreanu M, Niero R, Machado ID, Santin JR. Rubus imperialis (Rosaceae) extract and pure compound niga-ichigoside F1: wound healing and anti-inflammatory effects. Naunyn Schmiedebergs Arch Pharmacol 2016; 389:1235-1244. [PMID: 27527496 DOI: 10.1007/s00210-016-1285-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 08/08/2016] [Indexed: 12/13/2022]
Abstract
Here, we evaluate the anti-inflammatory and wound-healing effects of methanolic crude extract obtained from aerial parts (leaves and branches) of Rubus imperialis Chum. Schl. (Rosaceae) and the pure compound niga-ichigoside F1. Anti-inflammatory activity was determined in vivo and in vitro, and the healing effect was evaluated in surgical lesions in mice skin. The 1,1-diphenyl-2-picrylhydrazyl radical (DPPH) assay and H2O2-induced oxidative stress were used to determine antioxidant activity. The efferocytosis activity was also determined. The data obtained show that the extract of R. imperialis promote reduction in the inflammatory process induced by lipopolysaccharide (LPS) or carrageenan in the air pouch model; the effects could be reinforced by nitric oxide reduction in LPS-stimulated neutrophils, and an increase in the efferocytosis. The extract showed wound healing property in vitro and in vivo, scavenging activity for DPPH, and cytoprotection in the H2O2-induced oxidative stress in L929 cells. In addition, the compound niga-ichigoside F1 was able to reduce the NO secretion; however, it did not present wound-healing activity in vitro. Together, the data obtained point out the modulatory actions of R. imperialis extract on leukocyte migration to the inflamed tissue, the antioxidant, and the pro-resolutive activity. However, the R. imperialis anti-inflammatory activity may be mediated in parts by niga-ichigoside F1, and on wound healing do not correlated with niga-ichigoside F1.
Collapse
Affiliation(s)
- Talita Dacroce Tonin
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Liliani Carolini Thiesen
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Maria Luisa de Oliveira Nunes
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Milena Fronza Broering
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Marcos Paulo Donato
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Marina Jagielski Goss
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Marcel Petreanu
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Rivaldo Niero
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - Isabel Daufenback Machado
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil
| | - José Roberto Santin
- Programa de Pós-Graduação em Ciências Farmacêuticas and Núcleo de Investigações Químico-Farmacêuticas-NIQFAR, Universidade do Vale do Itajaí - UNIVALI, Rua Uruguai, 458, Itajaí, SC, 88302-901, Brazil.
| |
Collapse
|
31
|
Korner G, Scherer T, Adamsen D, Rebuffat A, Crabtree M, Rassi A, Scavelli R, Homma D, Ledermann B, Konrad D, Ichinose H, Wolfrum C, Horsch M, Rathkolb B, Klingenspor M, Beckers J, Wolf E, Gailus-Durner V, Fuchs H, Hrabě de Angelis M, Blau N, Rozman J, Thöny B. Mildly compromised tetrahydrobiopterin cofactor biosynthesis due to Pts variants leads to unusual body fat distribution and abdominal obesity in mice. J Inherit Metab Dis 2016; 39:309-19. [PMID: 26830550 DOI: 10.1007/s10545-015-9909-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 12/04/2015] [Accepted: 12/04/2015] [Indexed: 12/31/2022]
Abstract
Tetrahydrobiopterin (BH4) is an essential cofactor for the aromatic amino acid hydroxylases, alkylglycerol monooxygenase, and nitric oxide synthases (NOS). Inborn errors of BH4 metabolism lead to severe insufficiency of brain monoamine neurotransmitters while augmentation of BH4 by supplementation or stimulation of its biosynthesis is thought to ameliorate endothelial NOS (eNOS) dysfunction, to protect from (cardio-) vascular disease and/or prevent obesity and development of the metabolic syndrome. We have previously reported that homozygous knock-out mice for the 6-pyruvolytetrahydropterin synthase (PTPS; Pts-ko/ko) mice with no BH4 biosynthesis die after birth. Here we generated a Pts-knock-in (Pts-ki) allele expressing the murine PTPS-p.Arg15Cys with low residual activity (15% of wild-type in vitro) and investigated homozygous (Pts-ki/ki) and compound heterozygous (Pts-ki/ko) mutants. All mice showed normal viability and depending on the severity of the Pts alleles exhibited up to 90% reduction of PTPS activity concomitant with neopterin elevation and mild reduction of total biopterin while blood L-phenylalanine and brain monoamine neurotransmitters were unaffected. Yet, adult mutant mice with compromised PTPS activity (i.e., Pts-ki/ko, Pts-ki/ki or Pts-ko/wt) had increased body weight and elevated intra-abdominal fat. Comprehensive phenotyping of Pts-ki/ki mice revealed alterations in energy metabolism with proportionally higher fat content but lower lean mass, and increased blood glucose and cholesterol. Transcriptome analysis indicated changes in glucose and lipid metabolism. Furthermore, differentially expressed genes associated with obesity, weight loss, hepatic steatosis, and insulin sensitivity were consistent with the observed phenotypic alterations. We conclude that reduced PTPS activity concomitant with mildly compromised BH4-biosynthesis leads to abnormal body fat distribution and abdominal obesity at least in mice. This study associates a novel single gene mutation with monogenic forms of obesity.
Collapse
Affiliation(s)
- Germaine Korner
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
- Children's Research Center (CRC), Zürich, Switzerland
| | - Tanja Scherer
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
- Children's Research Center (CRC), Zürich, Switzerland
| | - Dea Adamsen
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland
- Children's Research Center (CRC), Zürich, Switzerland
| | - Alexander Rebuffat
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
| | - Mark Crabtree
- BHF Centre of Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, OX3 9DU, Oxford, UK
| | - Anahita Rassi
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zürich, Zürich, Switzerland
| | - Rossana Scavelli
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland
| | - Daigo Homma
- Department of Life Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Birgit Ledermann
- Division of Animal Facility, University of Zurich, Zürich, Switzerland
| | - Daniel Konrad
- Division of Pediatric Endocrinology and Diabetology, University Children's Hospital Zürich, Zürich, Switzerland
| | - Hiroshi Ichinose
- Department of Life Science, Graduate School of Bioscience and Biotechnology, Tokyo Institute of Technology, Yokohama, Japan
| | - Christian Wolfrum
- Institute of Food Nutrition and Health, Swiss Federal Institute of Technology Zürich, Zürich, Switzerland
| | - Marion Horsch
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Birgit Rathkolb
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, Munich, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Martin Klingenspor
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, Am Forum 8, 85354, Freising-Weihenstephan, Germany
- ZIEL - Center for Nutrition and Food Sciences, Technische Universität München, D-85350, Freising, Germany
| | - Johannes Beckers
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, D-85354, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, Ludwig-Maximilians-Universität München, Feodor-Lynen-Str. 25, 81377, Munich, Germany
| | - Valérie Gailus-Durner
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Helmut Fuchs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
- Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, D-85354, Freising-Weihenstephan, Germany
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland.
- Dietmar-Hopp Metabolic Center, University Children's Hospital Heidelberg, Im Neuenheimer Feld 669, D-69120, Heidelberg, Germany.
| | - Jan Rozman
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany.
- Molecular Nutritional Medicine, Else Kröner-Fresenius Center, Technische Universität München, Am Forum 8, 85354, Freising-Weihenstephan, Germany.
- German Center for Diabetes Research (DZD), Ingolstädter Landstrasse 1, 85764, Neuherberg, Germany.
| | - Beat Thöny
- Division of Metabolism, University Children's Hospital Zürich, Steinwiesstrasse 75, CH-8032, Zürich, Switzerland.
- Affiliated with the Neuroscience Center Zurich (ZNZ), University of Zurich and ETH Zurich, Zürich, Switzerland.
- Children's Research Center (CRC), Zürich, Switzerland.
| |
Collapse
|
32
|
Tillery LC, Epperson TA, Eguchi S, Motley ED. Featured Article: Differential regulation of endothelial nitric oxide synthase phosphorylation by protease-activated receptors in adult human endothelial cells. Exp Biol Med (Maywood) 2016; 241:569-80. [PMID: 26729042 DOI: 10.1177/1535370215622584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2015] [Accepted: 11/06/2015] [Indexed: 11/17/2022] Open
Abstract
Protease-activated receptors have been shown to regulate endothelial nitric oxide synthase through the phosphorylation of specific sites on the enzyme. It has been established that PAR-2 activation phosphorylates eNOS-Ser-1177 and leads to the production of the potent vasodilator nitric oxide, while PAR-1 activation phosphorylates eNOS-Thr-495 and decreases nitric oxide production in human umbilical vein endothelial cells. In this study, we hypothesize a differential coupling of protease-activated receptors to the signaling pathways that regulates endothelial nitric oxide synthase and nitric oxide production in primary adult human coronary artery endothelial cells. Using Western Blot analysis, we showed that thrombin and the PAR-1 activating peptide, TFLLR, lead to the phosphorylation of eNOS-Ser-1177 in human coronary artery endothelial cells, which was blocked by SCH-79797 (SCH), a PAR-1 inhibitor. Using the nitrate/nitrite assay, we also demonstrated that the thrombin- and TFLLR-induced production of nitric oxide was inhibited by SCH and L-NAME, a NOS inhibitor. In addition, we observed that TFLLR, unlike thrombin, significantly phosphorylated eNOS-Thr-495, which may explain the observed delay in nitric oxide production in comparison to that of thrombin. Activation of PAR-2 by SLIGRL, a PAR-2 specific ligand, leads to dual phosphorylation of both catalytic sites but primarily regulated eNOS-Thr-495 phosphorylation with no change in nitric oxide production in human coronary artery endothelial cells. PAR-3, known as the non-signaling receptor, was activated by TFRGAP, a PAR-3 mimicking peptide, and significantly induced the phosphorylation of eNOS-Thr-495 with minimal phosphorylation of eNOS-Ser-1177 with no change in nitric oxide production. In addition, we confirmed that PAR-mediated eNOS-Ser-1177 phosphorylation was Ca(2+)-dependent using the Ca(2+) chelator, BAPTA, while eNOS-Thr-495 phosphorylation was mediated via Rho kinase using the ROCK inhibitor, Y-27632, suggesting protease-activated receptor coupling to Gq and G12/13, respectively. These data suggest a vascular bed specific differential coupling of protease-activated receptors to the signaling pathways that regulate endothelial nitric oxide synthase and nitric oxide production that may be responsible for endothelial dysfunction associated with cardiovascular disease.
Collapse
Affiliation(s)
- Lakeisha C Tillery
- Department of Microbiology and Immunology, Meharry Medical College, Nashville, TN 37208, USA
| | - Tenille A Epperson
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Satoru Eguchi
- Department of Physiology, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Evangeline D Motley
- Department of Physiology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
33
|
Eriksson L, Nyström T. Antidiabetic agents and endothelial dysfunction - beyond glucose control. Basic Clin Pharmacol Toxicol 2015; 117:15-25. [PMID: 25827165 DOI: 10.1111/bcpt.12402] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 03/24/2015] [Indexed: 12/15/2022]
Abstract
Diabetes is rapidly increasing worldwide, and the number of patients suffering from diabetes is projected to rise by 50% over the next 25 years, then affecting almost 600 million adults. Type 2 diabetes comprises 90-95% of all people with diabetes, and they constitute a patient group that carries a high burden of cardiovascular disease. The relationship between hyperglycaemia and macrovascular complications is still uncertain, at least in terms of the possibility of reducing cardiovascular events solely by improving glycaemic control. This MiniReview has thus focused on the effect of common antidiabetic agents, with emphasis on glucagon-like peptide-1, on the endothelial cells of the vasculature. Patients with type 2 diabetes suffer a two to four times higher risk of myocardial infarction and stroke than healthy persons. In addition to this, patients with diabetes have an increased atherosclerotic burden. Endothelial dysfunction is thought to be an early and important predictor of atherosclerosis and cardiovascular disease, and in people with type 2 diabetes, endothelial dysfunction is a common finding. It is therefore of importance to consider whether drugs used within the clinical management of Type 2 diabetes mellitus (T2DM) exert direct and positive effects on the vasculature independent of their glucose-lowering ability. This might serve to limit the adverse consequences of the macrovascular complications of T2DM, as dysfunction of endothelial cells is believed to contribute to a premature development of atherosclerosis.
Collapse
Affiliation(s)
- Linnéa Eriksson
- Division of Vascular Surgery, Department of Molecular Medicine and Surgery, Centre for Molecular Medicine, Karolinska Institutet, Solna, Sweden
| | - Thomas Nyström
- Department of Clinical Science and Education, Karolinska Institutet, Stockholm South Hospital, Stockholm, Sweden
| |
Collapse
|
34
|
High-throughput intracellular pteridinic profiling by liquid chromatography–quadrupole time-of-flight mass spectrometry. Anal Chim Acta 2015; 853:442-450. [DOI: 10.1016/j.aca.2014.10.044] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 02/08/2023]
|
35
|
Ryan BJ, Crabtree MJ, Channon KM, Wade-Martins R. Parkinson's disease in GTP cyclohydrolase 1 mutation carriers. Brain 2014; 138:e348. [PMID: 25398233 DOI: 10.1093/brain/awu308] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Brent J Ryan
- 1 Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Mark J Crabtree
- 1 Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Keith M Channon
- 1 Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK 1 Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| | - Richard Wade-Martins
- 1 Oxford Parkinson's Disease Centre, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford OX1 3QX, UK
| |
Collapse
|
36
|
Ma S, Ma CCH. Recent developments in the effects of nitric oxide-donating statins on cardiovascular disease through regulation of tetrahydrobiopterin and nitric oxide. Vascul Pharmacol 2014; 63:63-70. [PMID: 25139660 DOI: 10.1016/j.vph.2014.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2014] [Revised: 08/01/2014] [Accepted: 08/04/2014] [Indexed: 10/24/2022]
Abstract
Since the discovery of the importance of nitric oxide (NO) to the human body three decades ago, numerous laboratory and clinical studies have been done to explore its potential therapeutic actions on many organs. In the cardiovascular system, NO works as a volatile signaling molecule regulating the vascular permeability and vascular tone, preventing thrombosis and inflammation, as well as inhibiting the smooth muscle hyperplasia. Thus, NO is important in the prevention and treatment of cardiovascular disease. NO is synthesized by NO synthase (NOS) with tetrahydrobiopterin (BH4) as the crucial cofactor. Many studies have been done to form nitric oxide donors so as to deliver NO directly to the vessel walls. In addition, NO moieties have been incorporated into existing therapeutic agents to enhance the NO bioavailability, including statins. Statins are inhibitors of 3-hydroxy-3-methylglutaryl-coenzyme (HMG-CoA), the rate-limiting enzyme of the mevalonate pathway. By inhibiting this pathway, statins lower blood cholesterol and exert their pleiotropic effects through activity in reaction cascades, such as Rho/ROCK and Rac 1/NADPH oxidase pathways. Statins have also been observed to implement their non-lipid effects by promoting BH4 synthesis with increase of NO bioavailability. Furthermore, NO-donating statins in laboratory studies have demonstrated to produce better therapeutic effects than their parent's drugs. They offer better anti-inflammatory, anti-proliferative and antithrombotic actions on cardiovascular system. They also cause better revascularization in peripheral ischemia and produce greater enhancement in limb reperfusion and salvage. In addition, it has been shown that NO-donating statin caused less myotoxicity, the most common side effect related to treatment with statins. The initial studies have demonstrated the superior therapeutic effects of NO-donating statins while producing fewer side effects.
Collapse
Affiliation(s)
- Sze Ma
- Hong Kong Baptist Hospital, Hong Kong; National University Ireland, Ireland; Royal College of Physicians of Ireland, Ireland
| | - Christopher Cheng-Hwa Ma
- NHS Dumfries & Galloway, GMC 7411692, United Kingdom; King's College London School of Medicine, United Kingdom.
| |
Collapse
|
37
|
Rojo AI, McBean G, Cindric M, Egea J, López MG, Rada P, Zarkovic N, Cuadrado A. Redox control of microglial function: molecular mechanisms and functional significance. Antioxid Redox Signal 2014; 21:1766-801. [PMID: 24597893 PMCID: PMC4186766 DOI: 10.1089/ars.2013.5745] [Citation(s) in RCA: 249] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neurodegenerative diseases are characterized by chronic microglial over-activation and oxidative stress. It is now beginning to be recognized that reactive oxygen species (ROS) produced by either microglia or the surrounding environment not only impact neurons but also modulate microglial activity. In this review, we first analyze the hallmarks of pro-inflammatory and anti-inflammatory phenotypes of microglia and their regulation by ROS. Then, we consider the production of reactive oxygen and nitrogen species by NADPH oxidases and nitric oxide synthases and the new findings that also indicate an essential role of glutathione (γ-glutamyl-l-cysteinylglycine) in redox homeostasis of microglia. The effect of oxidant modification of macromolecules on signaling is analyzed at the level of oxidized lipid by-products and sulfhydryl modification of microglial proteins. Redox signaling has a profound impact on two transcription factors that modulate microglial fate, nuclear factor kappa-light-chain-enhancer of activated B cells, and nuclear factor (erythroid-derived 2)-like 2, master regulators of the pro-inflammatory and antioxidant responses of microglia, respectively. The relevance of these proteins in the modulation of microglial activity and the interplay between them will be evaluated. Finally, the relevance of ROS in altering blood brain barrier permeability is discussed. Recent examples of the importance of these findings in the onset or progression of neurodegenerative diseases are also discussed. This review should provide a profound insight into the role of redox homeostasis in microglial activity and help in the identification of new promising targets to control neuroinflammation through redox control of the brain.
Collapse
Affiliation(s)
- Ana I Rojo
- 1 Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Berka V, Liu W, Wu G, Tsai AL. Comparison of oxygen-induced radical intermediates in iNOS oxygenase domain with those from nNOS and eNOS. J Inorg Biochem 2014; 139:93-105. [PMID: 25016313 DOI: 10.1016/j.jinorgbio.2014.06.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 06/17/2014] [Accepted: 06/17/2014] [Indexed: 12/31/2022]
Abstract
Inducible nitric-oxide synthase (iNOS) produces the reactive oxygen and nitrogen species (ROS/RNS) involved in bacteria killing and is crucial in the host defense mechanism. However, high level ROS/RNS can also be detrimental to normal cells and thus their production has to be tightly controlled. Availability or deficiency of tetrahydrobiopterin (BH4) cofactor and l-arginine substrate controls coupling or uncoupling of NOS catalysis. Fully coupled reaction, with abundant BH4 and l-arginine, produces NO whereas the uncoupled NOS (in the absence of BH4 and/or l-arginine) generates ROS/RNS. In the current work we focus on direct rapid freeze EPR to characterize the structure and kinetics of oxygen-induced radical intermediates produced by ferrous inducible NOS oxygenase domain (iNOSox) in the presence or absence of BH4 and/or l-arginine. Fully reconstituted iNOSox (+BH4, +L-Arg) forms a dimer and yields a typical BH4 radical that indicates coupled reaction. iNOSox (-BH4) remains mainly monomeric and produces exclusively superoxide, that is only marginally affected by the presence of l-arginine. iNOSox (+BH4, -L-Arg) exists as a monomer/dimer mixture and yields both BH4 radical and superoxide. Present study is a natural extension of our previous work on the ferrous endothelial NOSox (eNOSox) [V. Berka, G. Wu, H.C. Yeh, G. Palmer, A.L. Tsai, J. Biol. Chem. 279 (2004) 32243-32251] and ferrous neuronal NOSox (nNOSox) [V. Berka, L.H. Wang, A.L. Tsai, Biochemistry 47 (2008) 405-420]. Overall, our data suggests different regulatory roles of l-arginine and BH4 in the production of oxygen-induced radical intermediates in NOS isoforms which nicely serve individual functional role.
Collapse
Affiliation(s)
- Vladimír Berka
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States.
| | - Wen Liu
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Gang Wu
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Ah-Lim Tsai
- Division of Hematology, Department of Internal Medicine, University of Texas Health Science Center at Houston, Houston, TX 77030, United States.
| |
Collapse
|
39
|
Tveden-Nyborg P, Lykkesfeldt J. Does vitamin C deficiency increase lifestyle-associated vascular disease progression? Evidence based on experimental and clinical studies. Antioxid Redox Signal 2013; 19:2084-104. [PMID: 23642093 DOI: 10.1089/ars.2013.5382] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Despite continuous advances in the prevention of cardiovascular disease (CVD), critical issues associated with an unhealthy lifestyle remain an increasing cause of morbidity and mortality in industrialized countries. RECENT ADVANCES A growing body of literature supports a specific role for vitamin C in a number of reactions that are associated with vascular function and control including, for example, nitric oxide bioavailability, lipid metabolism, and vascular integrity. CRITICAL ISSUES A large body of epidemiological evidence supports a relationship between poor vitamin C status and increased risk of developing CVD, and the prevalence of deficiency continues to be around 10%-20% of the general Western population although this problem could easily and cheaply be solved by supplementation. However, large intervention studies using vitamin C have not found a beneficial effect of supplementation. This review outlines the proposed mechanism by which vitamin C deficiency worsens CVD progression. In addition, it discusses problems with the currently available literature, including the discrepancies between the large intervention studies and the experimental and epidemiological literature. FUTURE DIRECTIONS Increased insights into vitamin C deficiency-mediated CVD progression will enable the design of future randomized controlled trials that are better suited to test the efficacy of vitamin C in disease prevention as well as the identification of high-risk individuals which could possibly benefit from supplementation.
Collapse
Affiliation(s)
- Pernille Tveden-Nyborg
- Department of Veterinary Disease Biology, Faculty of Health and Medical Sciences, University of Copenhagen , Frederiksberg, Denmark
| | | |
Collapse
|
40
|
Liang Y, Inagaki H, Hao Q, Sakamoto M, Ohye T, Suzuki T, Ichinose H. Identification of an enhancer region for immune activation in the human GTP cyclohydrolase I gene. Biochem Biophys Res Commun 2013; 442:72-8. [DOI: 10.1016/j.bbrc.2013.11.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/02/2013] [Indexed: 11/16/2022]
|
41
|
Kar S, Kavdia M. Endothelial NO and O₂·⁻ production rates differentially regulate oxidative, nitroxidative, and nitrosative stress in the microcirculation. Free Radic Biol Med 2013; 63:161-74. [PMID: 23639567 PMCID: PMC4051226 DOI: 10.1016/j.freeradbiomed.2013.04.024] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2012] [Revised: 04/04/2013] [Accepted: 04/13/2013] [Indexed: 02/07/2023]
Abstract
Endothelial dysfunction causes an imbalance in endothelial NO and O₂·⁻ production rates and increased peroxynitrite formation. Peroxynitrite and its decomposition products cause multiple deleterious effects including tyrosine nitration of proteins, superoxide dismutase (SOD) inactivation, and tissue damage. Studies have shown that peroxynitrite formation during endothelial dysfunction is strongly dependent on the NO and O₂·⁻ production rates. Previous experimental and modeling studies examining the role of NO and O₂·⁻ production imbalance on peroxynitrite formation showed different results in biological and synthetic systems. However, there is a lack of quantitative information about the formation and biological relevance of peroxynitrite under oxidative, nitroxidative, and nitrosative stress conditions in the microcirculation. We developed a computational biotransport model to examine the role of endothelial NO and O₂·⁻ production on the complex biochemical NO and O₂·⁻ interactions in the microcirculation. We also modeled the effect of variability in SOD expression and activity during oxidative stress. The results showed that peroxynitrite concentration increased with increase in either O₂·⁻ to NO or NO to O₂·⁻ production rate ratio (QO₂·⁻/QNO or QNO/QO₂·⁻, respectively). The peroxynitrite concentrations were similar for both production rate ratios, indicating that peroxynitrite-related nitroxidative and nitrosative stresses may be similar in endothelial dysfunction or inducible NO synthase (iNOS)-induced NO production. The endothelial peroxynitrite concentration increased with increase in both QO₂·⁻/QNO and QNO/QO₂·⁻ ratios at SOD concentrations of 0.1-100 μM. The absence of SOD may not mitigate the extent of peroxynitrite-mediated toxicity, as we predicted an insignificant increase in peroxynitrite levels beyond QO₂·⁻/QNO and QNO/QO₂·⁻ ratios of 1. The results support the experimental observations of biological systems and show that peroxynitrite formation increases with increase in either NO or O₂·⁻ production, and excess NO production from iNOS or from NO donors during oxidative stress conditions does not reduce the extent of peroxynitrite mediated toxicity.
Collapse
Affiliation(s)
- Saptarshi Kar
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA.
| | | |
Collapse
|
42
|
Dulak J, Guzik TJ. Angiogenesis, stem cells, eNOS and inflammation--the many faces of vascular biology. Thromb Haemost 2012; 108:801-3. [PMID: 23052221 DOI: 10.1160/th12-10-0729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2012] [Accepted: 10/04/2012] [Indexed: 11/05/2022]
|