1
|
Kiesworo K, Agius T, Macarthur MR, Lambelet M, Lyon A, Zhang J, Turiel G, Fan Z, d’Almeida S, Uygun K, Yeh H, Déglise S, de Bock K, Mitchell SJ, Ocampo A, Allagnat F, Longchamp A. Nicotinamide mononucleotide restores impaired metabolism, endothelial cell proliferation and angiogenesis in old sedentary male mice. iScience 2025; 28:111656. [PMID: 39868046 PMCID: PMC11763620 DOI: 10.1016/j.isci.2024.111656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/15/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Aging is accompanied by a decline in neovascularization potential and increased susceptibility to ischemic injury. Here, we confirm the age-related impaired neovascularization following ischemic leg injury and impaired angiogenesis. The age-related deficits in angiogenesis arose primarily from diminished EC proliferation capacity, but not migration or VEGF sensitivity. Aged EC harvested from the mouse skeletal muscle displayed a pro-angiogenic gene expression phenotype, along with considerable changes in metabolic genes. Metabolomics analysis and 13C glucose tracing revealed impaired ATP production and blockade in glycolysis and TCA cycle in late passage HUVECs, which occurred at nicotinamide adenine dinucleotide (NAD⁺)-dependent steps, along with NAD+ depletion. Supplementation with nicotinamide mononucleotide (NMN), a precursor of NAD⁺, enhances late-passage EC proliferation and sprouting angiogenesis from aged mice aortas. Taken together, our study illustrates the importance of NAD+-dependent metabolism in the maintenance of EC proliferation capacity with age, and the therapeutic potential of NAD precursors.
Collapse
Affiliation(s)
- Kevin Kiesworo
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Michael R. Macarthur
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Martine Lambelet
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Arnaud Lyon
- Transplantation Centre and Transplantation Immunopathology Laboratory, Department of Medicine, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Jing Zhang
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Guillermo Turiel
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Zheng Fan
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | | | - Korkut Uygun
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Heidi Yeh
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sébastien Déglise
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Katrien de Bock
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Sarah J. Mitchell
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
- Ludwig Princeton Branch, Princeton University, Princeton, NJ, USA
| | - Alejandro Ocampo
- Department of Biomedical Sciences, Lausanne University (UNIL), Lausanne, Switzerland
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
2
|
Israel A, Raz I, Green I, Golan-Cohen A, Berkovitch M, Magen E, Vinker S, Merzon E. Health disparities in diabetes treatment: The challenge of G6PD deficiency. Diabetes Res Clin Pract 2025; 219:111965. [PMID: 39710070 DOI: 10.1016/j.diabres.2024.111965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
AIMS To assess the impact of Glucose-6-phosphate dehydrogenase (G6PD) deficiency, an enzymatic deficiency prevalent in individuals of African or Asian descent, on Hemoglobin A1c (HbA1c) levels, diabetes medication purchases, and the cumulative incidence of diabetes related complications. METHODS A large cohort study was conducted within a national health organization, comparing 3,913 G6PD-deficient patients to a matched control group without G6PD deficiency over two decades. The main measures and outcomes were the HbA1c levels, patterns of diabetes medication purchases, and the incidence of severe diabetes-related complications. RESULTS HbA1c levels significantly underestimated blood glucose concentrations in G6PD-deficient individuals. Individuals with diabetes and G6PD deficiency had lower rates of treatment with most diabetes medications, notably GLP-1 receptor agonists and SGLT2 inhibitors. Severe diabetes-related complications were more frequent among G6PD-deficient patients, with adjusted hazards ratios [95% confidence intervals] of 1.44 [1.16-1.81] for severe kidney insufficiency, 1.75 [1.23-2.49] for myocardial infarction, and 1.27 [1.02-1.58] for neuropathy. CONCLUSIONS This research highlights serious gaps in the management of G6PD-deficient patients with diabetes, who suffer from insufficient medication management and higher rates of complications. These findings underscore the need to account for G6PD deficiency in diabetes treatment to ensure equitable and effective healthcare for this vulnerable population.
Collapse
Affiliation(s)
- Ariel Israel
- Leumit Research Institute, Leumit Health Services, Tel-Aviv, Israel; Department of Epidemiology and Preventive Medicine, School of Public Health, Faculty of Medical & Health Sciences, Tel Aviv University, Tel-Aviv, Israel.
| | - Itamar Raz
- Diabetes Unit, Hadassah Medical Center, Jerusalem, Israel
| | - Ilan Green
- Leumit Research Institute, Leumit Health Services, Tel-Aviv, Israel; Department of Family Medicine, Faculty of Medical & Health Sciences, Tel-Aviv University, Israel
| | - Avivit Golan-Cohen
- Leumit Research Institute, Leumit Health Services, Tel-Aviv, Israel; Department of Family Medicine, Faculty of Medical & Health Sciences, Tel-Aviv University, Israel
| | - Matitiahu Berkovitch
- Clinical Pharmacology Unit, Shamir Medical Center, The Andy-Lebach Chair of Clinical Pharmacology and Toxicology, Medical School, Tel-Aviv University, Israel
| | - Eli Magen
- Medicine A Department, Assuta Ashdod University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Shlomo Vinker
- Leumit Research Institute, Leumit Health Services, Tel-Aviv, Israel; Department of Family Medicine, Faculty of Medical & Health Sciences, Tel-Aviv University, Israel
| | - Eugene Merzon
- Leumit Research Institute, Leumit Health Services, Tel-Aviv, Israel; Adelson School of Medicine, Ariel University, Ariel, Israel
| |
Collapse
|
3
|
Citrin KM, Chaube B, Fernández-Hernando C, Suárez Y. Intracellular endothelial cell metabolism in vascular function and dysfunction. Trends Endocrinol Metab 2024:S1043-2760(24)00296-0. [PMID: 39672762 DOI: 10.1016/j.tem.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/04/2024] [Accepted: 11/11/2024] [Indexed: 12/15/2024]
Abstract
Endothelial cells (ECs) form the inner lining of blood vessels that is crucial for vascular function and homeostasis. They regulate vascular tone, oxidative stress, and permeability. Dysfunction leads to increased permeability, leukocyte adhesion, and thrombosis. ECs undergo metabolic changes in conditions such as wound healing, cancer, atherosclerosis, and diabetes, and can influence disease progression. We discuss recent research that has revealed diverse intracellular metabolic pathways in ECs that are tailored to their functional needs, including lipid handling, glycolysis, and fatty acid oxidation (FAO). Understanding EC metabolic signatures in health and disease will be crucial not only for basic biology but can also be exploited when designing new therapies to target EC-related functions in different vascular diseases.
Collapse
Affiliation(s)
- Kathryn M Citrin
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Department of Cellular and Molecular Physiology, Yale University, New Haven, CT, USA
| | - Balkrishna Chaube
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Indian Institute of Technology Dharwad, Karnataka, India
| | - Carlos Fernández-Hernando
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yajaira Suárez
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA; Vascular Biology and Therapeutics Program, Yale University School of Medicine, New Haven, CT, USA; Yale Center for Molecular and System Metabolism, Yale University School of Medicine, New Haven, CT, USA; Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
4
|
Ma Y, Yuan X, Wei A, Li X, Patar A, Su S, Wang S, Ma G, Zhu J, Kong E. Enhancing Gpx1 palmitoylation to inhibit angiogenesis by targeting PPT1. Redox Biol 2024; 77:103376. [PMID: 39423458 PMCID: PMC11532489 DOI: 10.1016/j.redox.2024.103376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 09/28/2024] [Indexed: 10/21/2024] Open
Abstract
The significance of protein S-palmitoylation in angiogenesis has been largely overlooked, leaving various aspects unexplored. Recent identification of Gpx1 as a palmitoylated protein has generated interest in exploring its potential involvement in novel pathological mechanisms related to angiogenesis. In this study, we demonstrate that Gpx1 undergoes palmitoylation at cysteine-76 and -113, with PPT1 playing a crucial role in modulating the depalmitoylation of Gpx1. Furthermore, we find that PPT1-regulated depalmitoylation negatively impacts Gpx1 protein stability. Interestingly, inhibiting Gpx1 palmitoylation, either through expression of a non-palmitoylated Gpx1 mutant or by expressing PPT1, significantly enhances neovascular angiogenesis. Conversely, in PPT1-deficient mice, angiogenesis is notably attenuated compared to wild-type mice in an Oxygen-Induced Retinopathy (OIR) model, which mimics pathological angiogenesis. Physiologically, under hypoxic conditions, Gpx1 palmitoylation levels are drastically reduced, suggesting that increasing Gpx1 palmitoylation may have beneficial effects. Indeed, enhancing Gpx1 palmitoylation by inhibiting PPT1 with DC661 effectively suppresses retinal angiogenesis in the OIR disease model. Overall, our findings highlight the pivotal role of protein palmitoylation in angiogenesis and propose a novel mechanism whereby the PPT1-Gpx1 axis modulates angiogenesis, thereby providing a potential therapeutic strategy for targeting PPT1 to combat angiogenesis.
Collapse
Affiliation(s)
- Yidan Ma
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China; Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Xinxin Yuan
- Sanquan College of Xinxiang Medical University, XinXiang 453003, Henan, China
| | - Aodong Wei
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Xiaopeng Li
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Azim Patar
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, 16150, Malaysia
| | - Shaobo Su
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Songtao Wang
- Department of Human Anatomy & Histoembryology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, Henan, 453000, China
| | - Gaoen Ma
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China; The First Affiliated Hospital of Hainan Medical University, Haikou, 571199, China.
| | - Jiangli Zhu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China; Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Xinxiang Medical University, Xinxiang, 453000, China.
| | - Eryan Kong
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, 453000, China; Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Henan Health Commission Key Laboratory of Gastrointestinal Cancer Prevention and Treatment, Xinxiang Medical University, Xinxiang, 453000, China.
| |
Collapse
|
5
|
Xiao W, Lee LY, Loscalzo J. Metabolic Responses to Redox Stress in Vascular Cells. Antioxid Redox Signal 2024; 41:793-817. [PMID: 38985660 DOI: 10.1089/ars.2023.0476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
Significance: Redox stress underlies numerous vascular disease mechanisms. Metabolic adaptability is essential for vascular cells to preserve energy and redox homeostasis. Recent Advances: Single-cell technologies and multiomic studies demonstrate significant metabolic heterogeneity among vascular cells in health and disease. Increasing evidence shows that reductive or oxidative stress can induce metabolic reprogramming of vascular cells. A recent example is intracellular L-2-hydroxyglutarate accumulation in response to hypoxic reductive stress, which attenuates the glucose flux through glycolysis and mitochondrial respiration in pulmonary vascular cells and provides protection against further reductive stress. Critical Issues: Regulation of cellular redox homeostasis is highly compartmentalized and complex. Vascular cells rely on multiple metabolic pathways, but the precise connectivity among these pathways and their regulatory mechanisms is only partially defined. There is also a critical need to understand better the cross-regulatory mechanisms between the redox system and metabolic pathways as perturbations in either systems or their cross talk can be detrimental. Future Directions: Future studies are needed to define further how multiple metabolic pathways are wired in vascular cells individually and as a network of closely intertwined processes given that a perturbation in one metabolic compartment often affects others. There also needs to be a comprehensive understanding of how different types of redox perturbations are sensed by and regulate different cellular metabolic pathways with specific attention to subcellular compartmentalization. Lastly, integration of dynamic changes occurring in multiple metabolic pathways and their cross talk with the redox system is an important goal in this multiomics era. Antioxid. Redox Signal. 41,793-817.
Collapse
Affiliation(s)
- Wusheng Xiao
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
- Department of Toxicology, School of Public Health, Peking University, Beijing, China
| | - Laurel Y Lee
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Han B, Zhang Y, Liu C, Ji P, Xing Z, Geng X, Chi K, Gong M, Li Y, Zhang Y, Fu Z, Hong Q, Cai G, Chen X, Sun X. Renal inflammation combined with renal function reserve reduction accelerate kidney aging via pentose phosphate pathway. iScience 2024; 27:110045. [PMID: 38947529 PMCID: PMC11214290 DOI: 10.1016/j.isci.2024.110045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/20/2024] [Accepted: 05/17/2024] [Indexed: 07/02/2024] Open
Abstract
Aging is closely associated with inflammation, which affects renal function reserve (RFR) in the kidneys. This study aims to investigate the impact of reduced RFR reduction on kidney aging and the influence of renal inflammation and RFR reduction on this process. Natural aging rats and those subjected to unilateral nephrectomy (UNX), 1/6 nephrectomy (1/6NX), and unilateral ureteral obstruction (UUO) were observed at 6, 12, 18, and 21 months. Our findings suggest that RFR reduction and renal inflammation can accelerate kidney aging, and inflammation contributes more. Metabolomics analysis revealed alterations in amino acid metabolism contribute to RFR decline. Furthermore, experiments in vitro confirmed the involvement of pentose phosphate pathway (PPP) in promoting aging though inflammation. Our research provides novel insights into for the mechanism of kidney aging and provides indirect support for clinical treatment decisions, such as addressing kidney inflammation, stones, or tumors that may necessitate partial or complete nephrectomy.
Collapse
Affiliation(s)
- Bing Han
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - YiXuan Zhang
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Chao Liu
- Department of Critical Care Medicine, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Pengcheng Ji
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Zenghui Xing
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiaodong Geng
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Kun Chi
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Ming Gong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Yingying Li
- Department of Ultrasound, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Ying Zhang
- Department of Ultrasound, First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Zhangning Fu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Quan Hong
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Guangyan Cai
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xiangmei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| | - Xuefeng Sun
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Nephrology Institute of the Chinese People’s Liberation Army, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Disease Research, Beijing 100853, China
| |
Collapse
|
7
|
Wāng Y, Han Y, Xu DX. Developmental impacts and toxicological hallmarks of silver nanoparticles across diverse biological models. ENVIRONMENTAL SCIENCE AND ECOTECHNOLOGY 2024; 19:100325. [PMID: 38046179 PMCID: PMC10692670 DOI: 10.1016/j.ese.2023.100325] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 12/05/2023]
Abstract
Silver nanoparticles (AgNPs), revered for their antimicrobial prowess, have become ubiquitous in a range of products, from biomedical equipment to food packaging. However, amidst their rising popularity, concerns loom over their possible detrimental effects on fetal development and subsequent adult life. This review delves into the developmental toxicity of AgNPs across diverse models, from aquatic species like zebrafish and catfish to mammalian rodents and in vitro embryonic stem cells. Our focus encompasses the fate of AgNPs in different contexts, elucidating associated hazardous results such as embryotoxicity and adverse pregnancy outcomes. Furthermore, we scrutinize the enduring adverse impacts on offspring, spanning impaired neurobehavior function, reproductive disorders, cardiopulmonary lesions, and hepatotoxicity. Key hallmarks of developmental harm are identified, encompassing redox imbalances, inflammatory cascades, DNA damage, and mitochondrial stress. Notably, we explore potential explanations, linking immunoregulatory dysfunction and disrupted epigenetic modifications to AgNPs-induced developmental failures. Despite substantial progress, our understanding of the developmental risks posed by AgNPs remains incomplete, underscoring the urgency of further research in this critical area.
Collapse
Affiliation(s)
- Yán Wāng
- Department of Toxicology, School of Public Health & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - Yapeng Han
- Department of Toxicology, School of Public Health & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| | - De-Xiang Xu
- Department of Toxicology, School of Public Health & Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
8
|
Karunakaran U, Elumalai S, Chung SM, Maedler K, Won KC, Moon JS. Mitochondrial aldehyde dehydrogenase-2 coordinates the hydrogen sulfide - AMPK axis to attenuate high glucose-induced pancreatic β-cell dysfunction by glutathione antioxidant system. Redox Biol 2024; 69:102994. [PMID: 38128451 PMCID: PMC10776427 DOI: 10.1016/j.redox.2023.102994] [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: 10/25/2023] [Revised: 12/05/2023] [Accepted: 12/09/2023] [Indexed: 12/23/2023] Open
Abstract
Progression of β-cell loss in diabetes mellitus is significantly influenced by persistent hyperglycemia. At the cellular level, a number of signaling cascades affect the expression of apoptotic genes, ultimately resulting in β-cell failure; these cascades have not been elucidated. Mitochondrial aldehyde dehydrogenase-2 (ALDH2) plays a central role in the detoxification of reactive aldehydes generated from endogenous and exogenous sources and protects against mitochondrial deterioration in cells. Here we report that under diabetogenic conditions, ALDH2 is strongly inactivated in β-cells through CDK5-dependent glutathione antioxidant imbalance by glucose-6-phosphate dehydrogenase (G6PD) degradation. Intriguingly, CDK5 inhibition strengthens mitochondrial antioxidant defense through ALDH2 activation. Mitochondrial ALDH2 activation selectively preserves β-cells against high-glucose-induced dysfunction by activating AMPK and Hydrogen Sulfide (H2S) signaling. This is associated with the stabilization and enhancement of the activity of G6PD by SIRT2, a cytoplasmic NAD+-dependent deacetylase, and is thereby linked to an elevation in the GSH/GSSG ratio, which leads to the inhibition of mitochondrial dysfunction under high-glucose conditions. Furthermore, treatment with NaHS, an H2S donor, selectively preserves β-cell function by promoting ALDH2 activity, leading to the inhibition of lipid peroxidation by high-glucose concentrations. Collectively, our results provide the first direct evidence that ALDH2 activation enhances H2S-AMPK-G6PD signaling, leading to improved β-cell function and survival under high-glucose conditions via the glutathione redox balance.
Collapse
Affiliation(s)
- Udayakumar Karunakaran
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu, Republic of Korea.
| | - Suma Elumalai
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu, Republic of Korea
| | - Seung Min Chung
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu, Republic of Korea; Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea
| | - Kathrin Maedler
- Islet Biology Laboratory, Centre for Biomolecular Interactions Bremen, University of Bremen, Bremen, Germany
| | - Kyu Chang Won
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu, Republic of Korea; Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea.
| | - Jun Sung Moon
- Innovative Center for Aging Research, Yeungnam University Medical Center, Daegu, Republic of Korea; Department of Internal Medicine, Yeungnam University College of Medicine, Daegu, Republic of Korea.
| |
Collapse
|
9
|
Signoretti C, Gupte SA. G6PD Orchestrates Genome-Wide DNA Methylation and Gene Expression in the Vascular Wall. Int J Mol Sci 2023; 24:16727. [PMID: 38069050 PMCID: PMC10706803 DOI: 10.3390/ijms242316727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/22/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Recent advances have revealed the importance of epigenetic modifications to gene regulation and transcriptional activity. DNA methylation, a determinant of genetic imprinting and the de novo silencing of genes genome-wide, is known to be controlled by DNA methyltransferases (DNMT) and demethylases (TET) under disease conditions. However, the mechanism(s)/factor(s) influencing the expression and activity of epigenetic writers and erasers, and thus DNA methylation, in healthy vascular tissue is incompletely understood. Based on our recent studies, we hypothesized that glucose-6-phosphate dehydrogenase (G6PD) is a modifier of DNMT and TET expression and activity and an enabler of gene expression. In the aorta of CRISPR-edited rats with the Mediterranean G6PD variant, we determined DNA methylation by whole-genome bisulfite sequencing, gene expression by RNA sequencing, and large artery stiffness by echocardiography. Here, we documented higher expression of Dnmt1, Dnmt3a, Tet2, and Tet3 in aortas from Mediterranean G6PDS188F variant (a loss-of-function single nucleotide polymorphism) rats than their wild-type littermates. Concomitantly, we identified 17,618 differentially methylated loci genome-wide (5787 hypermethylated loci, including down-regulated genes encoding inflammation- and vasoconstriction-causing proteins, and 11,827 hypomethylated loci, including up-regulated genes encoding smooth muscle cell differentiation- and fatty acid metabolism-promoting proteins) in aortas from G6PDS188F as compared to wild-type rats. Our results demonstrated that nitric oxide, which is generated in a G6PD-derived NADPH-dependent manner, increases TET and decreases DNMT activity. Further, we observed less large artery (aorta) stiffness in G6PDS188F as compared to wild-type rats. These results establish a noncanonical function of the wild-type G6PD and G6PDS188F variant in the regulation of DNA methylation and gene expression in healthy vascular tissue and reveal that the G6PDS188F variant contributes to reducing large artery stiffness.
Collapse
Affiliation(s)
| | - Sachin A. Gupte
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA;
| |
Collapse
|
10
|
Moraes B, Martins R, Lopes C, Martins R, Arcanjo A, Nascimento J, Konnai S, da Silva Vaz I, Logullo C. G6PDH as a key immunometabolic and redox trigger in arthropods. Front Physiol 2023; 14:1287090. [PMID: 38046951 PMCID: PMC10693429 DOI: 10.3389/fphys.2023.1287090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 10/30/2023] [Indexed: 12/05/2023] Open
Abstract
The enzyme glucose-6-phosphate dehydrogenase (G6PDH) plays crucial roles in glucose homeostasis and the pentose phosphate pathway (PPP), being also involved in redox metabolism. The PPP is an important metabolic pathway that produces ribose and nicotinamide adenine dinucleotide phosphate (NADPH), which are essential for several physiologic and biochemical processes, such as the synthesis of fatty acids and nucleic acids. As a rate-limiting step in PPP, G6PDH is a highly conserved enzyme and its deficiency can lead to severe consequences for the organism, in particular for cell growth. Insufficient G6PDH activity can lead to cell growth arrest, impaired embryonic development, as well as a reduction in insulin sensitivity, inflammation, diabetes, and hypertension. While research on G6PDH and PPP has historically focused on mammalian models, particularly human disorders, recent studies have shed light on the regulation of this enzyme in arthropods, where new functions were discovered. This review will discuss the role of arthropod G6PDH in regulating redox homeostasis and immunometabolism and explore potential avenues for further research on this enzyme in various metabolic adaptations.
Collapse
Affiliation(s)
- Bruno Moraes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Renato Martins
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Cintia Lopes
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Ronald Martins
- Programa de Computação Científica, Instituto Oswaldo Cruz, IOC, Rio de Janeiro, Brazil
| | - Angélica Arcanjo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Jhenifer Nascimento
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| | - Satoru Konnai
- Laboratory of Infectious Diseases, Hokkaido University, Sapporo, Japan
| | - Itabajara da Silva Vaz
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
- Centro de Biotecnologia and Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Carlos Logullo
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Entomologia Molecular–INCT, Rio de Janeiro, Brazil
| |
Collapse
|
11
|
Dow LF, Case AM, Paustian MP, Pinkerton BR, Simeon P, Trippier PC. The evolution of small molecule enzyme activators. RSC Med Chem 2023; 14:2206-2230. [PMID: 37974956 PMCID: PMC10650962 DOI: 10.1039/d3md00399j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/20/2023] [Indexed: 11/19/2023] Open
Abstract
There is a myriad of enzymes within the body responsible for maintaining homeostasis by providing the means to convert substrates to products as and when required. Physiological enzymes are tightly controlled by many signaling pathways and their products subsequently control other pathways. Traditionally, most drug discovery efforts focus on identifying enzyme inhibitors, due to upregulation being prevalent in many diseases and the existence of endogenous substrates that can be modified to afford inhibitor compounds. As enzyme downregulation and reduction of endogenous activators are observed in multiple diseases, the identification of small molecules with the ability to activate enzymes has recently entered the medicinal chemistry toolbox to afford chemical probes and potential therapeutics as an alternative means to intervene in diseases. In this review we highlight the progress made in the identification and advancement of non-kinase enzyme activators and their potential in treating various disease states.
Collapse
Affiliation(s)
- Louise F Dow
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Alfie M Case
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Megan P Paustian
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Braeden R Pinkerton
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Princess Simeon
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center Omaha NE 68106 USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center Omaha NE 68106 USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center Omaha NE 68106 USA
| |
Collapse
|
12
|
Wiśniewski K, Popęda M, Price B, Bieńkowski M, Fahlström A, Drummond K, Adamides AA. Glucose-6-phosphate dehydrogenase and 8-iso-prostaglandin F2α as potential predictors of delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage. J Neurosurg 2023; 139:698-707. [PMID: 36640097 DOI: 10.3171/2022.12.jns222332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/07/2022] [Indexed: 01/15/2023]
Abstract
OBJECTIVE Delayed cerebral ischemia (DCI) is a serious complication of aneurysmal subarachnoid hemorrhage (aSAH), which is responsible for significant death and disability. The dynamic balance between the production and elimination of reactive oxygen species (ROS) in patients with DCI is suspected be shifted to favor ROS formation. The authors assessed the relationship between F2-isoprostanes (F2-IsoPs), oxidative stress biomarkers, and glucose-6-phosphate dehydrogenase (G6PD), which are responsible for nicotinamide adenine dinucleotide phosphate (NADPH) production for glutathione system function, with post-aSAH DCI. METHODS The authors assessed 45 aSAH patients for F2-IsoP and G6PD concentration using commercial ELISA on days 2, 4, and 6 after aSAH. The authors examined the correlation between plasma F2-IsoP and G6PD concentrations and clinical factors with DCI occurrence and aSAH outcome. RESULTS Expectedly, the most important clinical predictors of DCI were Hunt and Hess grade and modified Fisher (mFisher) grade. Plasma F2-IsoP and G6PD concentrations were greater in aSAH patients than the control group (p < 0.01). F2-IsoP concentrations were greater and G6PD concentrations were lower in patients with DCI than those without (p < 0.01). Plasma F2-IsoP and G6PD concentrations on day 2 were correlated with DCI occurrence (p < 0.01). Plasma F2-IsoP concentrations on days 2 and 6 were correlated with outcome at 1 and 12 months (p < 0.01). CONCLUSIONS Decreased G6PD indirectly informs the reduced antioxidant response, especially for the glutathione system. G6PD concentration was lower in patients with DCI than those without, which may explain the increased F2-IsoP concentrations. mFisher grade, plasma F2-IsoP concentration, and G6PD concentration on day 2 after aSAH, in combination, may serve as predictors of DCI. Further research is necessary to investigate the therapeutic utility of F2-IsoPs and antioxidants in clinical practice.
Collapse
Affiliation(s)
- Karol Wiśniewski
- 1Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- 2Department of Neurosurgery and Neurooncology, Medical University of Łódź, Łódzkie, Poland
| | - Marta Popęda
- 3Department of Pathomorphology, Medical University of Gdańsk, Pomorskie, Poland
| | - Benjamin Price
- 1Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Michał Bieńkowski
- 3Department of Pathomorphology, Medical University of Gdańsk, Pomorskie, Poland
| | - Andreas Fahlström
- 1Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- 4Department of Medical Sciences, Section of Neurosurgery, Uppsala University, Uppsala, Sweden; and
| | - Katharine Drummond
- 1Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- 5Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Victoria, Australia
| | - Alexios A Adamides
- 1Department of Neurosurgery, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- 5Department of Surgery, Royal Melbourne Hospital, University of Melbourne, Victoria, Australia
| |
Collapse
|
13
|
Manzano-Pech L, Guarner-Lans V, Elena Soto M, Díaz-Díaz E, Pérez-Torres I. Alteration of the aortic vascular reactivity associated to excessive consumption of Hibiscus sabdariffa Linnaeus: Preliminary findings. Heliyon 2023; 9:e20020. [PMID: 37810051 PMCID: PMC10559753 DOI: 10.1016/j.heliyon.2023.e20020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 08/18/2023] [Accepted: 09/08/2023] [Indexed: 10/10/2023] Open
Abstract
The moderate production of reactive oxidative species (ROS) is important because ROS act as second messengers. However, their depletion through the over-activity of the antioxidant system may lead to reductive stress (RS) which is characterized by an increase in reducing equivalents and an elevation of some components of the antioxidant system disturbing redox homeostasis. Hibiscus sabdariffa Linnaeus (HSL) is a plant with antioxidant properties that provides compounds that favor the antioxidant system. However, excess chronic consumption could lead to the over expression of the antioxidant enzymatic system, and this could contribute to decrease ROS. Therefore, the objective of this study was to evaluate the alteration of the vascular reactivity associated to excessive and chronic consumption of HSL infusions at different percentages. 40 male Wistar rats were divided into 4 groups. Group 1 control (drinking tap water), group 2, 3 and 4, drinking water supplemented with 15, 30 and 60 g/L of HSL calyxes respectively. The systolic blood pressure (SBP), vascular reactivity, morphological changes, and different components of the enzymatic antioxidant system were evaluated in the thoracic aorta by spectrophotometry. We also determined glucose-6-phosphate dehydrogenase (G6PD), glutathione-S-transferase (GST), thioredoxin-reductase (TrxR), glutathione peroxidase (GPx) and glutathione reductase (GR) and some markers of the non-enzimatic system such as the NO3-/NO2-ratio, glutathione (GSH), selenium, thiols, lipoperoxidation (LPO), and 3-nitrityrosine (3-NT). Vasoconstriction was increased and vasorelaxation was decreased. These alterations were reversed by O2- and H2O2. There was an increase in the wall thickness and elastic fibers (p = 0.004 and p = 0.02, respectively) and in G6PD, GPX, TrxR (p = 0.02, p = 0.03, and p = 0.01 respectively). LPO, GSH (p = 0.01), and selenium (p = 0.04) were decreased. There was a decrease in thiols (p < 0.001), 3-NT (p = 0.04) and GST (p = 0.0005) in rats that received the infusion at 3 and 6%. The excess antioxidants provided by the HSL infusions at 3% and 6% modified vascular reactivity, increasing the enzymatic antioxidant system, and depleting ROS.
Collapse
Affiliation(s)
- Linaloe Manzano-Pech
- Departments of Cardiovascular Biomedicine, Institute National of Cardiology Ignacio Chávez, Juan Badiano 1, Section XVI, Tlalpan, 14080, México, USA
| | - Verónica Guarner-Lans
- Physiology Institute National of Cardiology Ignacio Chávez, Juan Badiano 1, Section XVI, Tlalpan 14080, México, USA
| | - María Elena Soto
- Immunology, Institute National of Cardiology Ignacio Chávez, Juan Badiano 1, Section XVI, Tlalpan, 14080, México, USA
| | - Eulises Díaz-Díaz
- Department of Reproductive Biology, Institute National of medical Sciences and Nutrition Salvador Zubirán, Vasco de Quiroga 15, Section XVI, Tlalpan, 14000, México, USA
| | - Israel Pérez-Torres
- Departments of Cardiovascular Biomedicine, Institute National of Cardiology Ignacio Chávez, Juan Badiano 1, Section XVI, Tlalpan, 14080, México, USA
| |
Collapse
|
14
|
Zaied AA, Ushio‐Fukai M, Fukai T, Kovacs‐Kasa A, Alhusban S, Sudhahar V, Ganta VC, Annex BH. Pentose Pathway Activation Is Superior to Increased Glycolysis for Therapeutic Angiogenesis in Peripheral Arterial Disease. J Am Heart Assoc 2023; 12:e027986. [PMID: 36974760 PMCID: PMC10122893 DOI: 10.1161/jaha.122.027986] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/05/2022] [Indexed: 03/29/2023]
Abstract
Background In endothelial cells (ECs), glycolysis, regulated by PFKFB3 (6-phosphofructo-2-kinase/fructose-2,6-biphosphatase, isoform-3), is the major metabolic pathway for ATP generation. In preclinical peripheral artery disease models, VEGF165a (vascular endothelial growth factor165a) and microRNA-93 both promote angiogenesis. Methods and Results Mice following hind-limb ischemia (HLI) and ECs with, and without, hypoxia and serum starvation were examined with, and without, microRNA-93 and VEGF165a. Post-HLI perfusion recovery was monitored. EC metabolism was studied using seahorse assay, and the expression and activity of major metabolism genes were assessed. Reactive oxygen species levels and EC permeability were evaluated. C57Bl/6J mice generated a robust angiogenic response to HLI, with ECs from ischemic versus nonischemic muscle demonstrating no increase in glycolysis. Balb/CJ mice generated a poor angiogenic response post-HLI; ischemic versus nonischemic ECs demonstrated significant increase in glycolysis. MicroRNA-93-treated Balb/CJ mice post-HLI showed better perfusion recovery, with ischemic versus nonischemic ECs showing no increase in glycolysis. VEGF165a-treated Balb/CJ mice post-HLI showed no improvement in perfusion recovery with ischemic versus nonischemic ECs showing significant increase in glycolysis. ECs under hypoxia and serum starvation upregulated PFKFB3. In ECs under hypoxia and serum starvation, VEGF165a versus control significantly upregulated PFKFB3 and glycolysis, whereas miR-93 versus control demonstrated no increase in PFKFB3 or glycolysis. MicroRNA-93 versus VEGF165a upregulated glucose-6-phosphate dehydrogenase expression and activity, activating the pentose phosphate pathway. MicroRNA-93 versus control increased reduced nicotinamide adenine dinucleotide phosphate and virtually eliminated the increase in reactive oxygen species. In ECs under hypoxia and serum starvation, VEGF165a significantly increased and miR-93 decreased EC permeability. Conclusions In peripheral artery disease, activation of the pentose phosphate pathway to promote angiogenesis may offer potential therapeutic advantages.
Collapse
Affiliation(s)
- Abdelrahman A. Zaied
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Masuko Ushio‐Fukai
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Tohru Fukai
- Departments of Pharmacology and ToxicologyMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Charlie Norwood Veterans Affairs Medical CenterAugustaGAUSA
| | - Anita Kovacs‐Kasa
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Suhib Alhusban
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Varadarajan Sudhahar
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Vijay C. Ganta
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
| | - Brian H. Annex
- Vascular Biology CenterMedical College of Georgia at Augusta UniversityAugustaGAUSA
- Department of MedicineMedical College of Georgia at Augusta UniversityAugustaGAUSA
| |
Collapse
|
15
|
Abdelghffar EAR, ALmohammadi AG, Malik S, Khalphallah A, Soliman MM. Changes in clinicomorphometrical findings, lipid profiles, hepatorenal indices and oxidant/antioxidant status as thermoregulatory adaptive mechanisms in poikilothermic Dabb lizard (Uromastyx aegyptia). Sci Rep 2023; 13:3409. [PMID: 36854728 PMCID: PMC9975223 DOI: 10.1038/s41598-023-30184-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 02/17/2023] [Indexed: 03/02/2023] Open
Abstract
Wildlife has exposed to various environmental stressors. Reptiles (ectothermic) are highly susceptible to climatic changes due to their behaviour, physiology, and life history that were so heavily reliant on the ambient environmental temperature. The present work aims to monitor different biochemical and haematological indices of Dabb lizards (Uromastyx aegyptia) at various thermal gradients as well as their adaptation to oxidative stress. This has been reflected through assessment of their impact on some adaptive physiological traits i.e. thermoregulation, and muscle metabolic biomarkers, blood pictures and oxidant/antioxidant status. This experiment is carried out on non-hibernating adult male Dabb lizards (U. aegyptia; n = 24) of age of 18-24 months. These Dabb lizards are divided into four equal groups (n = 6 for each one) where they are exposed to different thermal treatments for one week as following; control group [Exposed to terrarium temperature 38-39 °C], low temperature exposed group [Exposed to 12-14 °C], Gp. C; moderate temperature exposed group [Exposed to 41-43 °C] and high temperature exposed group [Exposed to 43-45 °C]. Each independent group (n = 6) are kept at separated glass terraria. The investigated lizards are monitored for body temperature, morphometric measurements i.e. body weight (g) and total body length (cm; TBL), muscle biochemical analysis, haematological pictures indices and serum biochemical assays including mainly oxidant/antioxidants biomarkers throughout the current experiment. The results state that the thermoregulatory behaviour of Dabb varies with the increase of concentration of muscular metabolic enzymes. In low temperature exposed group, the increase in red blood corpuscles (RBCs), haemoglobin concentrations (Hb), white blood cell (WBC), serum antioxidant biomarkers and anaerobic Lactate dehydrogenase (LDH) enzyme are associated with a marked reduction in serum levels of total cholesterol (TC), triglycerides (TGs), total proteins (TPs), albumin, glucose and electrolytes. In moderate temperature exposed group, a significant elevation in serum values of TC, TGs, TPs, glucose, urea and uric acids levels are mentioned. In high temperature exposed Dabb group, a remarkable increase in blood values of RBCs, Hb, haematocrit value (HCT), WBC, T. chol., TGs, TPs, glucose, urea, uric acids, triiodothyronine (T3) and thyroxine (T4) levels are also observed. Moreover, significant increases in muscular anaerobic/aerobic metabolic enzymes as well as stimulation of antioxidant defence system have been reported. Different significant correlations have been stated between variably estimated laboratory indices in the investigated Dabb lizards under different thermal treatments. The study concludes that the Dabb lizards have a strong antioxidant defence system and undergo physiological thermoregulatory adaptive mechanisms, that involve biochemical and metabolic acclimatization as a response to environmental temperature changes that act as a protective mechanism against oxidative stress as well as maintained homeostatic responses and normal physiological functions.
Collapse
Affiliation(s)
- Eman A R Abdelghffar
- Department of Zoology, Faculty of Science, Ain Shams University, Abbasseya, Cairo, 11566, Egypt.
- Department of Biology, College of Science, Taibah University, Yanbu, Kingdom of Saudi Arabia.
| | - Ameera G ALmohammadi
- Department of Biology, College of Science, Taibah University, Yanbu, Kingdom of Saudi Arabia
| | - Samina Malik
- University College of Medicine and Dentistry, University of Lahore, Lahore, Pakistan
| | - Arafat Khalphallah
- Division of Internal Medicine, Department of Animal Medicine, Faculty of Veterinary Medicine, Assiut University, Assiut, 71526, Egypt.
| | - Mohamed Mostafa Soliman
- Department of Zoology, Faculty of Science, Ain Shams University, Abbasseya, Cairo, 11566, Egypt
- Biology Department, Faculty of Science, Jazan University, Jazan, Kingdom of Saudi Arabia
| |
Collapse
|
16
|
Azouaoui D, Choinière MR, Khan M, Sayfi S, Jaffer S, Yousef S, Patten DA, Green AE, Menzies KJ. Meta-analysis of NAD(P)(H) quantification results exhibits variability across mammalian tissues. Sci Rep 2023; 13:2464. [PMID: 36774401 PMCID: PMC9922293 DOI: 10.1038/s41598-023-29607-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 02/07/2023] [Indexed: 02/13/2023] Open
Abstract
Nicotinamide Adenine Dinucleotide (NAD+) plays an important role in energy metabolism and signaling pathways controlling crucial cellular functions. The increased interest in NAD+ metabolism and NAD+-boosting therapies has reinforced the necessity for accurate NAD+ quantification. To examine the published NAD(P)(H) measures across mammalian tissues, we performed a meta-analysis of the existing data. An Ovid MEDLINE database search identified articles with NAD(P)(H) quantification results obtained from mammalian tissues published between 1961 and 2021. We screened 4890 records and extracted quantitative data, as well as the quantification methods, pre-analytical conditions, and subject characteristics. The extracted physiological NAD(P)(H) concentrations in various tissues from mice, rats, and humans, revealed an important inter- and intra-method variability that extended to recent publications. This highlights the relatively poor potential for cross-experimental analyses for NAD(P)(H) quantitative data and the importance of standardization for NAD(P)(H) quantification methods and pre-analytical procedures for future preclinical and clinical studies.
Collapse
Affiliation(s)
- Dassine Azouaoui
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Michael René Choinière
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Momtafin Khan
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Shahab Sayfi
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Simran Jaffer
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Selvia Yousef
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - David A Patten
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Alexander E Green
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Keir J Menzies
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada.
- Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada.
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
17
|
Serna E, Mauricio MD, San-Miguel T, Guerra-Ojeda S, Verdú D, Valls A, Arc-Chagnaud C, De la Rosa A, Viña J. Glucose 6-P Dehydrogenase Overexpression Improves Aging-Induced Endothelial Dysfunction in Aorta from Mice: Role of Arginase II. Int J Mol Sci 2023; 24:ijms24043622. [PMID: 36835034 PMCID: PMC9961129 DOI: 10.3390/ijms24043622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
The increase of vascular arginase activity during aging causes endothelial dysfunction. This enzyme competes with the endothelial nitric oxide synthase (eNOS) for L-arginine substrate. Our hypothesis is that glucose 6-P dehydrogenase (G6PD) overexpression could improve the endothelial function modulating the arginase pathway in aorta from mice. For this study, three groups of male mice were used: young wild type (WT) (6-9 months), old WT (21-22 months) and old G6PD-Tg (21-22 months) mice. Vascular reactivity results showed a reduced acetylcholine-dependent relaxation in the old WT but not old G6PD-Tg group. Endothelial dysfunction was reverted by nor-NOHA, an arginase inhibitor. Mice overexpressing G6PD underexpressed arginase II and also displayed a lower activity of this enzyme. Moreover, histological analyses demonstrated that age causes a thickness of aortic walls, but this did not occur in G6PD-Tg mice. We conclude that the overexpressing G6PD mouse is a model to improve vascular health via the arginase pathway.
Collapse
Affiliation(s)
- Eva Serna
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
- Correspondence:
| | - Maria D Mauricio
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Teresa San-Miguel
- Department of Pathology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Sol Guerra-Ojeda
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - David Verdú
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Alicia Valls
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Coralie Arc-Chagnaud
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - Adrián De la Rosa
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| | - José Viña
- Department of Physiology, Faculty of Medicine, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
18
|
Endothelial senescence in vascular diseases: current understanding and future opportunities in senotherapeutics. Exp Mol Med 2023; 55:1-12. [PMID: 36599934 PMCID: PMC9898542 DOI: 10.1038/s12276-022-00906-w] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 10/25/2022] [Accepted: 10/31/2022] [Indexed: 01/05/2023] Open
Abstract
Senescence compromises the essential role that the endothelium plays in maintaining vascular homeostasis, so promoting endothelial dysfunction and the development of age-related vascular diseases. Their biological and clinical significance calls for strategies for identifying and therapeutically targeting senescent endothelial cells. While senescence and endothelial dysfunction have been studied extensively, distinguishing what is distinctly endothelial senescence remains a barrier to overcome for an effective approach to addressing it. Here, we review the mechanisms underlying endothelial senescence and the evidence for its clinical importance. Furthermore, we discuss the current state and the limitations in the approaches for the detection and therapeutic intervention of target cells, suggesting potential directions for future research.
Collapse
|
19
|
Arasimowicz-Jelonek M, Jagodzik P, Płóciennik A, Sobieszczuk-Nowicka E, Mattoo A, Polcyn W, Floryszak-Wieczorek J. Dynamics of nitration during dark-induced leaf senescence in Arabidopsis reveals proteins modified by tryptophan nitration. JOURNAL OF EXPERIMENTAL BOTANY 2022; 73:6853-6875. [PMID: 35981877 DOI: 10.1093/jxb/erac341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 08/16/2022] [Indexed: 06/15/2023]
Abstract
Nitric oxide (NO) is a critical molecule that links plant development with stress responses. Herein, new insights into the role of NO metabolism during leaf senescence in Arabidopsis are presented. A gradual decrease in NO emission accompanied dark-induced leaf senescence (DILS), and a transient wave of peroxynitrite (ONOO-) formation was detected by day 3 of DILS. The boosted ONOO- did not promote tryptophan (Trp) nitration, while the pool of 6-nitroTrp-containing proteins was depleted as senescence progressed. Immunoprecipitation combined with mass spectrometry was used to identify 63 and 4 characteristic 6-nitroTrp-containing proteins in control and individually darkened leaves, respectively. The potential in vivo targets of Trp nitration were mainly related to protein biosynthesis and carbohydrate metabolism. In contrast, nitration of tyrosine-containing proteins was intensified 2-fold on day 3 of DILS. Also, nitrative modification of RNA and DNA increased significantly on days 3 and 7 of DILS, respectively. Taken together, ONOO- can be considered a novel pro-senescence regulator that fine-tunes the redox environment for selective bio-target nitration. Thus, DILS-triggered nitrative changes at RNA and protein levels promote developmental shifts during the plant's lifespan and temporal adjustment in plant metabolism under suboptimal environmental conditions.
Collapse
Affiliation(s)
- Magdalena Arasimowicz-Jelonek
- Department of Plant Ecophysiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Przemysław Jagodzik
- Department of Plant Ecophysiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Artur Płóciennik
- Department of Plant Ecophysiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Ewa Sobieszczuk-Nowicka
- Department of Plant Physiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | - Autar Mattoo
- Sustainable Agricultural Systems Laboratory, USDA-ARS, Henry A. Wallace Beltsville Agricultural Research Center, Beltsville, MD 20705-2350, USA
| | - Władysław Polcyn
- Department of Plant Physiology, Faculty of Biology, Adam Mickiewicz University; Uniwersytetu Poznańskiego 6, 61-614 Poznań, Poland
| | | |
Collapse
|
20
|
Yang C, Zhang X, Ge X, He C, Liu S, Yang S, Huang C. N-Acetylcysteine protects against cobalt chloride-induced endothelial dysfunction by enhancing glucose-6-phosphate dehydrogenase activity. FEBS Open Bio 2022; 12:1475-1488. [PMID: 35666067 PMCID: PMC9340863 DOI: 10.1002/2211-5463.13449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 05/05/2022] [Accepted: 06/03/2022] [Indexed: 11/07/2022] Open
Abstract
Hypoxia‐induced endothelial dysfunction is known to be involved in the pathogenesis of several vascular diseases. However, it remains unclear whether the pentose phosphate pathway (PPP) is involved in regulating the response of endothelial cells to hypoxia. Here, we established an in vitro model by treating EA.hy926 (a hybrid human umbilical vein cell line) with cobalt chloride (CoCl2; a chemical mimic that stabilizes HIF‐1α, thereby leading to the development of hypoxia), and used this to investigate the involvement of PPP by examining expression of its key enzyme, glucose‐6‐phosphate dehydrogenase (G6PD). We report that CoCl2 induces the accumulation of HIF‐1α, leading to endothelial cell dysfunction characterized by reduced cell viability, proliferation, tube formation, and activation of cytokine production, accompanied with a significant decrease in G6PD expression and activity. The addition of 6‐aminonicotinamide (6‐AN) to inhibit PPP directly causes endothelial dysfunction. Additionally, N‐Acetylcysteine (NAC), a precursor of glutathione, was further evaluated for its protective effects; NAC displayed a protective effect against CoCl2‐induced cell damage by enhancing G6PD activity, and this was abrogated by 6‐AN. The effects of CoCl2 and the involvement of G6PD in endothelial dysfunction have been confirmed in primary human aortic endothelial cells. In summary, G6PD was identified as a novel target of CoCl2‐induced damage, which highlighted the involvement of PPP in regulating the response of endothelial cell CoCl2. Treatment with NAC may be a potential strategy to treat hypoxia or ischemia, which are widely observed in vascular diseases.
Collapse
Affiliation(s)
- Chen Yang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.,Department of Geriatrics, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Xiaofang Zhang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Xilin Ge
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Chunmei He
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Suhuan Liu
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.,Research Center for Translational Medicine, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Shuyu Yang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China.,Traditional Chinese Medicine research studio, the First Affiliated Hospital of Xiamen University, Xiamen, 361003, China
| | - Caoxin Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361003, China
| |
Collapse
|
21
|
Li J, Chen Y, Ou Z, Zhang Y, Liang Z, Deng W, Chen H, Huang W, He Y, Xing S, Yu J, Zeng J. Glucose-6-phosphate dehydrogenase deficiency and intracranial atherosclerotic stenosis in stroke patients. Eur J Neurol 2022; 29:2683-2689. [PMID: 35608963 DOI: 10.1111/ene.15418] [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: 02/11/2022] [Revised: 04/29/2022] [Accepted: 05/19/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND AND PURPOSE Intracranial atherosclerotic stenosis (ICAS) is a major cause of stroke in Asian countries. Glucose-6-phosphate dehydrogenase (G6PD) deficiency, a hereditary enzyme defect prevalent in Asian countries, has been associated with atherosclerotic cardiovascular disease and worse poststroke outcomes. However, the impact of G6PD deficiency on ICAS remains unclear. We aimed to compare the risk of ICAS in stroke patients with and without G6PD deficiency in a Chinese cohort. METHODS We prospectively and consecutively recruited stroke patients from four centers in China. All patients received intracranial artery assessment by magnetic resonance/computed tomography angiography or digital subtraction angiography, as well as G6PD enzyme evaluation. The prevalence, burden, and characteristics of ICAS were compared between patients with and without G6PD deficiency using multivariate regression analysis. RESULTS Among 1593 patients, 116 (63.7%) of 182 patients with G6PD deficiency and 714 (50.6%) of 1411 patients with normal G6PD levels were identified as ICAS. Age, hypertension, diabetes, and G6PD deficiency were independent predictors of ICAS. Among patients with ICAS, G6PD-deficient individuals were more likely to have multiple (≥2 segments) intracranial stenosis (odds ratio [OR] = 1.87, 95% confidence interval [CI] = 1.25-2.81, p = 0.002). G6PD deficiency increased the risk of ICAS in patients who were male (OR = 1.82, 95% CI = 1.24-2.66, p = 0.002), aged ≥70 years (OR = 2.40, 95% CI = 1.33-4.31, p = 0.004), or hypertensive (OR = 1.88, 95% CI = 1.28-2.77, p = 0.001). CONCLUSIONS Stroke patients with G6PD deficiency have a higher prevalence and ICAS burden than those with normal G6PD, particularly those who are male, older, and hypertensive.
Collapse
Affiliation(s)
- Jianle Li
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Yicong Chen
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Zilin Ou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Yusheng Zhang
- Department of Neurology, First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Zhijian Liang
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weisheng Deng
- Department of Neurology, Meizhou People's Hospital, Meizhou, China
| | - Hao Chen
- Department of Neurology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Weixian Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Yingxin He
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Shihui Xing
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Jian Yu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| | - Jinsheng Zeng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou, China
| |
Collapse
|
22
|
Costanzo-Garvey DL, Case AJ, Watson GF, Alsamraae M, Chatterjee A, Oberley-Deegan RE, Dutta S, Abdalla MY, Kielian T, Lindsey ML, Cook LM. Prostate cancer addiction to oxidative stress defines sensitivity to anti-tumor neutrophils. Clin Exp Metastasis 2022; 39:641-659. [PMID: 35604506 PMCID: PMC9338904 DOI: 10.1007/s10585-022-10170-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 04/24/2022] [Indexed: 01/17/2023]
Abstract
Bone metastatic prostate cancer (BM-PCa) remains one of the most difficult cancers to treat due to the complex interactions of cancer and stromal cells. We previously showed that bone marrow neutrophils elicit an anti-tumor immune response against BM-PCa. Further, we demonstrated that BM-PCa induces neutrophil oxidative burst, which has previously been identified to promote primary tumor growth of other cancers, and a goal of this study was to define the importance of neutrophil oxidative burst in BM-PCa. To do this, we first examined the impact of depletion of reactive oxygen species (ROS), via systemic deletion of the main source of ROS in phagocytes, NADPH oxidase (Nox)2, which we found to suppress prostate tumor growth in bone. Further, using pharmacologic ROS inhibitors and Nox2-null neutrophils, we found that ROS depletion specifically suppresses growth of androgen-insensitive prostate cancer cells. Upon closer examination using bulk RNA sequencing analysis, we identified that metastatic prostate cancer induces neutrophil transcriptomic changes that activates pathways associated with response to oxidative stress. In tandem, prostate cancer cells resist neutrophil anti-tumor response via extracellular (i.e., regulation of neutrophils) and intracellular alterations of glutathione synthesis, the most potent cellular antioxidant. These findings demonstrate that BM-PCa thrive under oxidative stress conditions and such that regulation of ROS and glutathione programming could be leveraged for targeting of BM-PCa progression.
Collapse
Affiliation(s)
- Diane L Costanzo-Garvey
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68198, USA
| | - Adam J Case
- Department of Psychiatry and Behavioral Sciences, Texas A&M College of Medicine, Bryan, TX, USA.,Department of Medical Physiology, Texas A&M College of Medicine, Bryan, TX, USA
| | - Gabrielle F Watson
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center and Omaha VA Medical Center, Omaha, NE, USA
| | - Massar Alsamraae
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68198, USA
| | - Arpita Chatterjee
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Rebecca E Oberley-Deegan
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Samikshan Dutta
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Maher Y Abdalla
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68198, USA
| | - Tammy Kielian
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68198, USA
| | - Merry L Lindsey
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center and Omaha VA Medical Center, Omaha, NE, USA
| | - Leah M Cook
- Department of Pathology and Microbiology, University of Nebraska Medical Center, 985900 Nebraska Med Center, Omaha, NE, 68198, USA.
| |
Collapse
|
23
|
Masih Uzza M, Khalilulla H, Osman Elha G, Mahmood T, Ahsan F, Karim S, Siddiqui NA, Ahamad SR, Alam Khan M, Khan A, Uzzaman Kh W, A.M. Abdul M, Ben Salah G. Anti-Diabetic Potential of Common Saudi Medicinal Herbs Commiphora molmol and Astragalus membranaceus Extracts in Diabetic Rats. INT J PHARMACOL 2022. [DOI: 10.3923/ijp.2022.475.487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
24
|
Neurons undergo pathogenic metabolic reprogramming in models of familial ALS. Mol Metab 2022; 60:101468. [PMID: 35248787 PMCID: PMC8958550 DOI: 10.1016/j.molmet.2022.101468] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 02/25/2022] [Accepted: 02/25/2022] [Indexed: 11/21/2022] Open
Abstract
Objectives Methods Results Conclusions Our work is the first to perform a comprehensive and quantitative analysis of intermediary metabolism in neurons in the setting of fALS causing gene products. Because the cardinal feature of ALS is death of motor neurons, these new studies are directly relevant to the pathogenesis of ALS. Our functional interrogations begin to unpack how metabolic re-wiring is induced by fALS genes and it will be very interesting, in the future, to gain insight in amino acid fueling of the TCA cycle. We suspect pleiotropic effects of amino acid fueling, and this may lead to very targeted therapeutic interventions.
Collapse
|
25
|
Li J, Sun Y, Liu F, Zhou Y, Yan Y, Zhou Z, Wang P, Zhou S. Increasing NADPH impairs fungal H 2O 2 resistance by perturbing transcriptional regulation of peroxiredoxin. BIORESOUR BIOPROCESS 2022; 9:1. [PMID: 38647831 PMCID: PMC10992141 DOI: 10.1186/s40643-021-00489-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 12/14/2021] [Indexed: 12/27/2022] Open
Abstract
NADPH provides the reducing power for decomposition of reactive oxygen species (ROS), making it an indispensable part during ROS defense. It remains uncertain, however, if living cells respond to the ROS challenge with an elevated intracellular NADPH level or a more complex NADPH-mediated manner. Herein, we employed a model fungus Aspergillus nidulans to probe this issue. A conditional expression of glucose-6-phosphate dehydrogenase (G6PD)-strain was constructed to manipulate intracellular NADPH levels. As expected, turning down the cellular NADPH concentration drastically lowered the ROS response of the strain; it was interesting to note that increasing NADPH levels also impaired fungal H2O2 resistance. Further analysis showed that excess NADPH promoted the assembly of the CCAAT-binding factor AnCF, which in turn suppressed NapA, a transcriptional activator of PrxA (the key NADPH-dependent ROS scavenger), leading to low antioxidant ability. In natural cell response to oxidative stress, we noticed that the intracellular NADPH level fluctuated "down then up" in the presence of H2O2. This might be the result of a co-action of the PrxA-dependent NADPH consumption and NADPH-dependent feedback of G6PD. The fluctuation of NADPH is well correlated to the formation of AnCF assembly and expression of NapA, thus modulating the ROS defense. Our research elucidated how A. nidulans precisely controls NADPH levels for ROS defense.
Collapse
Affiliation(s)
- Jingyi Li
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Yanwei Sun
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Feiyun Liu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Yao Zhou
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Yunfeng Yan
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhemin Zhou
- Key Laboratory of Industrial Biotechnology (Ministry of Education), School of Biotechnology, Jiangnan University, 1800 Lihu Avenue, Wuxi, 214122, Jiangsu, China
| | - Ping Wang
- Department of Bioproducts and Biosystems Engineering, University of Minnesota, Twin cities, Saint Paul, MN, 55108, USA.
| | - Shengmin Zhou
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
26
|
Lee LYH, Oldham WM, He H, Wang R, Mulhern R, Handy DE, Loscalzo J. Interferon-γ Impairs Human Coronary Artery Endothelial Glucose Metabolism by Tryptophan Catabolism and Activates Fatty Acid Oxidation. Circulation 2021; 144:1612-1628. [PMID: 34636650 DOI: 10.1161/circulationaha.121.053960] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Endothelial cells depend on glycolysis for much of their energy production. Impaired endothelial glycolysis has been associated with various vascular pathobiologies, including impaired angiogenesis and atherogenesis. IFN-γ (interferon-γ)-producing CD4+ and CD8+ T lymphocytes have been identified as the predominant pathological cell subsets in human atherosclerotic plaques. Although the immunologic consequences of these cells have been extensively evaluated, their IFN-γ-mediated metabolic effects on endothelial cells remain unknown. The purpose of this study was to determine the metabolic consequences of the T-lymphocyte cytokine, IFN-γ, on human coronary artery endothelial cells. METHODS The metabolic effects of IFN-γ on primary human coronary artery endothelial cells were assessed by unbiased transcriptomic and metabolomic analyses combined with real-time extracellular flux analyses and molecular mechanistic studies. Cellular phenotypic correlations were made by measuring altered endothelial intracellular cGMP content, wound-healing capacity, and adhesion molecule expression. RESULTS IFN-γ exposure inhibited basal glycolysis of quiescent primary human coronary artery endothelial cells by 20% through the global transcriptional suppression of glycolytic enzymes resulting from decreased basal HIF1α (hypoxia-inducible factor 1α) nuclear availability in normoxia. The decrease in HIF1α activity was a consequence of IFN-γ-induced tryptophan catabolism resulting in ARNT (aryl hydrocarbon receptor nuclear translocator)/HIF1β sequestration by the kynurenine-activated AHR (aryl hydrocarbon receptor). In addition, IFN-γ resulted in a 23% depletion of intracellular nicotinamide adenine dinucleotide in human coronary artery endothelial cells. This altered glucose metabolism was met with concomitant activation of fatty acid oxidation, which augmented its contribution to intracellular ATP balance by >20%. These metabolic derangements were associated with adverse endothelial phenotypic changes, including decreased basal intracellular cGMP, impaired endothelial migration, and a switch to a proinflammatory state. CONCLUSIONS IFN-γ impairs endothelial glucose metabolism by altered tryptophan catabolism destabilizing HIF1, depletes nicotinamide adenine dinucleotide, and results in a metabolic shift toward increased fatty acid oxidation. This work suggests a novel mechanistic basis for pathological T lymphocyte-endothelial interactions in atherosclerosis mediated by IFN-γ, linking endothelial glucose, tryptophan, and fatty acid metabolism with the nicotinamide adenine dinucleotide balance and ATP generation and their adverse endothelial functional consequences.
Collapse
Affiliation(s)
- Laurel Yong-Hwa Lee
- Division of Cardiovascular Medicine (L.Y.-H.L., H.H., R.W., R.M., D.E.H., J.L.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - William M Oldham
- Division of Pulmonary and Critical Care (W.M.O.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Huamei He
- Division of Cardiovascular Medicine (L.Y.-H.L., H.H., R.W., R.M., D.E.H., J.L.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Ruisheng Wang
- Division of Cardiovascular Medicine (L.Y.-H.L., H.H., R.W., R.M., D.E.H., J.L.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Ryan Mulhern
- Division of Cardiovascular Medicine (L.Y.-H.L., H.H., R.W., R.M., D.E.H., J.L.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Diane E Handy
- Division of Cardiovascular Medicine (L.Y.-H.L., H.H., R.W., R.M., D.E.H., J.L.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Joseph Loscalzo
- Division of Cardiovascular Medicine (L.Y.-H.L., H.H., R.W., R.M., D.E.H., J.L.), Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
27
|
Volkova N, Yuhta M, Sokil L, Chernyshenko L, Stepanuk L, Goltsev A. Efficiency of Combined Use of Fullerene C60 and Bovine Serum Albumin for Rehabilitation of Vitrified Fragments of Rat Immature Seminiferous Tubules. INNOVATIVE BIOSYSTEMS AND BIOENGINEERING 2021. [DOI: 10.20535/ibb.2021.5.3.241126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
Background. Today, cryopreserved reproductive tissues are used to treat some forms of male infertility. However, after long-term preservation of fragments of seminiferous tubules of testes (FSTT) in a low-temperature bank (-196 °С) their morphological and functional characteristics decrease reversibly. To solve this problem after freezing-thawing, the use of rehabilitation media with special additives is promising. Due to the fact that serum albumin and fullerene C60 have powerful protective and antioxidant properties, their use allows to stabilize the plasma membrane, osmotic pressure, and reduce free radicals that make them promising candidates to use in the development of rehabilitation media for biological objects after cryopreservation.
Objective. The efficacy of fullerene C60, bovine serum albumin (BSA), and their combination as components of rehabilitation medium of vitrified FSTT of immature rats was studied.
Methods. Vitrified-thawed samples of FSTT were incubated (22 °C) for 30 minutes in Leibovitz's medium with addition of 15 mg/mL C60, 5 g/L BSA or their combination. Control samples were incubated in the medium without C60 or BSA addition. Metabolic activity (MTT test), histomorphological data, total antioxidant status (TAS), reactive oxygen species (ROS) production, activity of g-glutamyltransferase (gGGT), and glucose-6-phosphate dehydrogenase were determined in the samples after rehabilitation in the investigated media.
Results. The use of C60 led to the increase of metabolic (by 1.26 times) and TAS (by 1.74 times) activities, to the decrease in the number of ROS+ cells (by 1.35 times) and to the improvement of the spermatogenic epithelium binding to the basement membrane versus control sample. Application of BSA did not significantly affect the studied biochemical indices but decreased the number of tubules with desquamation of spermatogenic epithelium in histological sections. The combined use of BSA and C60 had the best effect among investigated rehabilitation media that led to the increase of metabolic activity (by 1.51 times), TAS activity (by 1.78 times), gGGT activity (by 1.59 times), histostructure restoration and the decrease in the number of ROS+ cells (by 1.45 times) compared to the control samples.
Conclusions. The use of C60 and BSA combination increases the metabolic and antioxidant activity of vitrified FSTT and also has a positive effect on their histostructural characteristics compared to control samples. It should be noted that the effect of С60 and BSA addition to rehabilitation medium exceeds the results of using the investigated additives separately (by the metabolic and gGGT activity as well as architectonics of vitrified FSTT). These data relate to reproductive medicine and can be used to develop an effective rehabilitation protocol for vitrified FSTT.
Collapse
|
28
|
Abd El-Hameed AM, Yousef AI, Abd El-Twab SM, El-Shahawy AAG, Abdel-Moneim A. Hepatoprotective Effects of Polydatin-Loaded Chitosan Nanoparticles in Diabetic Rats: Modulation of Glucose Metabolism, Oxidative Stress, and Inflammation Biomarkers. BIOCHEMISTRY (MOSCOW) 2021; 86:179-189. [PMID: 33832416 DOI: 10.1134/s0006297921020061] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Polydatin (PD) has a broad range of pharmacological activities; however, its effects on diabetic liver damage are poorly studies. This work is aimed to explore possible protective effects of polydatin-loaded chitosan nanoparticles (PD-CSNPs) or PD against liver damage associated with diabetes. Diabetes was induced in rats using nicotinamide/streptozotocin treatment. Diabetic rats were then divided into six groups: normal control rats, diabetic control rats, and rats orally treated with PD, PD-CSNPs, equivalent unloaded CSNPs, or metformin daily for 4 weeks. Treatment with PD and PD-CSNPs significantly reduced the blood glucose content, lipid peroxidation in the liver, and activities of serum transaminases and carbohydrate metabolism enzymes (including succinate dehydrogenase and pyruvate kinase); by contrast, liver glycogen content, glutathione concentration, and activities of the antioxidant enzymes (superoxide dismutase, glutathione peroxidase, catalase, and glucose-6-phosphate dehydrogenase) were markedly increased compared with the control diabetic rats. Furthermore, expression of the tumor necrosis factor α and interleukin-1β mRNAs was significantly downregulated, while expression of glucose transporter 2 and glucokinase mRNAs was strongly upregulated vs. control diabetic rats. We concluded that PD-CSNPs and PD ameliorate diabetic liver damage by modulating glucose transporter 2 expression, affecting the activity of carbohydrate metabolism enzymes, and suppressing oxidative stress and inflammation, PD-CSNPs being more efficient than PD, probably due to higher bioavailability and prolonged release.
Collapse
Affiliation(s)
- Abeer M Abd El-Hameed
- Chemistry Department, Faculty of Science, Taibah University, Al-Madinah Al-Munawarah, 30002, Saudi Arabia.
| | - Ahmed I Yousef
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt.
| | - Sanaa M Abd El-Twab
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt.
| | - Ahmed A G El-Shahawy
- Materials Science and Nanotechnology Department, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, 62511, Egypt.
| | - Adel Abdel-Moneim
- Molecular Physiology Division, Faculty of Science, Beni-Suef University, Beni-Suef, 62511, Egypt.
| |
Collapse
|
29
|
Ushio-Fukai M, Ash D, Nagarkoti S, Belin de Chantemèle EJ, Fulton DJR, Fukai T. Interplay Between Reactive Oxygen/Reactive Nitrogen Species and Metabolism in Vascular Biology and Disease. Antioxid Redox Signal 2021; 34:1319-1354. [PMID: 33899493 PMCID: PMC8418449 DOI: 10.1089/ars.2020.8161] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Reactive oxygen species (ROS; e.g., superoxide [O2•-] and hydrogen peroxide [H2O2]) and reactive nitrogen species (RNS; e.g., nitric oxide [NO•]) at the physiological level function as signaling molecules that mediate many biological responses, including cell proliferation, migration, differentiation, and gene expression. By contrast, excess ROS/RNS, a consequence of dysregulated redox homeostasis, is a hallmark of cardiovascular disease. Accumulating evidence suggests that both ROS and RNS regulate various metabolic pathways and enzymes. Recent studies indicate that cells have mechanisms that fine-tune ROS/RNS levels by tight regulation of metabolic pathways, such as glycolysis and oxidative phosphorylation. The ROS/RNS-mediated inhibition of glycolytic pathways promotes metabolic reprogramming away from glycolytic flux toward the oxidative pentose phosphate pathway to generate nicotinamide adenine dinucleotide phosphate (NADPH) for antioxidant defense. This review summarizes our current knowledge of the mechanisms by which ROS/RNS regulate metabolic enzymes and cellular metabolism and how cellular metabolism influences redox homeostasis and the pathogenesis of disease. A full understanding of these mechanisms will be important for the development of new therapeutic strategies to treat diseases associated with dysregulated redox homeostasis and metabolism. Antioxid. Redox Signal. 34, 1319-1354.
Collapse
Affiliation(s)
- Masuko Ushio-Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Dipankar Ash
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Sheela Nagarkoti
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Eric J Belin de Chantemèle
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Medicine (Cardiology) and Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - David J R Fulton
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA
| | - Tohru Fukai
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Department of Pharmacology and Toxicology, Medical College of Georgia, Augusta University, Augusta, Georgia, USA.,Charlie Norwood Veterans Affairs Medical Center, Augusta, Georgia, USA
| |
Collapse
|
30
|
Badmus OO, Areola ED, Benjamin E, Obekpa MA, Adegoke TE, Elijah OE, Imam A, Olajide OJ, Olatunji LA. Suppression of Adenosine Deaminase and Xanthine Oxidase Activities by Mineralocorticoid and Glucocorticoid Receptor Blockades Restores Renal Antioxidative Barrier in Oral Contraceptive-Treated Dam. J Renin Angiotensin Aldosterone Syst 2021; 2021:9966372. [PMID: 34285713 PMCID: PMC8265027 DOI: 10.1155/2021/9966372] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/10/2021] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE We tested the hypothesis that postpartum combined oral contraceptive (COC) treatment would induce oxidative stress via the adenosine deaminase-xanthine oxidase pathway in the kidney. We also sought to determine whether mineralocorticoid receptor (MR) or glucocorticoid receptor (GR ) blockade would suppress the activities of ADA and xanthine oxidase caused by postpartum COC treatment in the kidney. METHODS Twenty-four Wistar dams were randomly assigned to 4 groups (n = 6/group). Dams received vehicle (po), COC (1.0 μg ethinylestradiol and 5.0 μg levonorgestrel; po), COC with GR blockade (mifepristone; 80.0 mg/kg; po), and COC with MR blockade (spironolactone; 0.25 mg/kg; po) daily between 3rd and 11th week postpartum. RESULTS Data showed that postpartum COC caused increased plasma creatinine and urea, increased renal triglyceride/high-density lipoprotein ratio, free fatty acid accumulation, alanine aminotransferase, gamma-glutamyltransferase, uric acid, and activities of renal XO and ADA. On the other hand, postpartum COC resulted in decreased plasma albumin, renal glutathione, and Na+-K+-ATPase activity with no effect on lactate production. However, MR or GR blockade ameliorated the alterations induced by postpartum COC treatment. The present results demonstrate that MR or GR blockade ameliorates postpartum COC-induced increased activities of ADA and xanthine oxidase and restores glutathione-dependent antioxidative defense. CONCLUSION These findings implicate the involvements of GR and MR in renal dysfunctions caused by COC in dams via disrupted glutathione antioxidative barrier.
Collapse
Affiliation(s)
- Olufunto O. Badmus
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Public Health, Kwara State University, Malete, Nigeria
| | - Emmanuel D. Areola
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Eleojo Benjamin
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Matthew A. Obekpa
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Tolulope E. Adegoke
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Oluwatobi E. Elijah
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Aminu Imam
- Department of Anatomy, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olayemi J. Olajide
- Department of Anatomy, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A. Olatunji
- HOPE Cardiometabolic Research Team and Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
31
|
Certo M, Elkafrawy H, Pucino V, Cucchi D, Cheung KC, Mauro C. Endothelial cell and T-cell crosstalk: Targeting metabolism as a therapeutic approach in chronic inflammation. Br J Pharmacol 2021; 178:2041-2059. [PMID: 31999357 PMCID: PMC8246814 DOI: 10.1111/bph.15002] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/09/2020] [Accepted: 01/15/2020] [Indexed: 12/14/2022] Open
Abstract
The role of metabolic reprogramming in the coordination of the immune response has gained increasing consideration in recent years. Indeed, it has become clear that changes in the metabolic status of immune cells can alter their functional properties. During inflammation, T cells need to generate sufficient energy and biomolecules to support growth, proliferation, and effector functions. Therefore, T cells need to rearrange their metabolism to meet these demands. A similar metabolic reprogramming has been described in endothelial cells, which have the ability to interact with and modulate the function of immune cells. In this overview, we will discuss recent insights in the complex crosstalk between endothelial cells and T cells as well as their metabolic reprogramming following activation. We highlight key components of this metabolic switch that can lead to the development of new therapeutics against chronic inflammatory disorders. LINKED ARTICLES: This article is part of a themed issue on Cellular metabolism and diseases. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.10/issuetoc.
Collapse
Affiliation(s)
- Michelangelo Certo
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Hagar Elkafrawy
- Medical Biochemistry and Molecular Biology Department, Faculty of MedicineAlexandria UniversityAlexandriaEgypt
| | - Valentina Pucino
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Danilo Cucchi
- Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Kenneth C.P. Cheung
- School of Life SciencesThe Chinese University of Hong KongHong Kong SARChina
| | - Claudio Mauro
- Institute of Inflammation and Ageing, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Cardiovascular Sciences, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
- Institute of Metabolism and Systems Research, College of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| |
Collapse
|
32
|
The Controversial Role of Glucose-6-Phosphate Dehydrogenase Deficiency on Cardiovascular Disease: A Narrative Review. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5529256. [PMID: 34007401 PMCID: PMC8110402 DOI: 10.1155/2021/5529256] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 03/27/2021] [Accepted: 04/21/2021] [Indexed: 12/12/2022]
Abstract
Cardiovascular disorders (CVD) are highly prevalent and the leading cause of death worldwide. Atherosclerosis is responsible for most cases of CVD. The plaque formation and subsequent thrombosis in atherosclerosis constitute an ongoing process that is influenced by numerous risk factors such as hypertension, diabetes, dyslipidemia, obesity, smoking, inflammation, and sedentary lifestyle. Among the various risk and protective factors, the role of glucose-6-phosphate dehydrogenase (G6PD) deficiency, the most common inborn enzyme disorder across populations, is still debated. For decades, it has been considered a protective factor against the development of CVD. However, in the recent years, growing scientific evidence has suggested that this inherited condition may act as a CVD risk factor. The role of G6PD deficiency in the atherogenic process has been investigated using in vitro or ex vivo cellular models, animal models, and epidemiological studies in human cohorts of variable size and across different ethnic groups, with conflicting results. In this review, the impact of G6PD deficiency on CVD was critically reconsidered, taking into account the most recent acquisitions on molecular and biochemical mechanisms, namely, antioxidative mechanisms, glutathione recycling, and nitric oxide production, as well as their mutual interactions, which may be impaired by the enzyme defect in the context of the pentose phosphate pathway. Overall, current evidence supports the notion that G6PD downregulation may favor the onset and evolution of atheroma in subjects at risk of CVD. Given the relatively high frequency of this enzyme deficiency in several regions of the world, this finding might be of practical importance to tailor surveillance guidelines and facilitate risk stratification.
Collapse
|
33
|
Alagbonsi AI, Salman TM, Sulaiman SO, Adedini KA, Kebu S. Possible mechanisms of the hypoglycaemic effect of artesunate: Gender implication. Metabol Open 2021; 10:100087. [PMID: 33778463 PMCID: PMC7985403 DOI: 10.1016/j.metop.2021.100087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 03/08/2021] [Indexed: 02/07/2023] Open
Abstract
Background We investigated the mechanism of artesunate’s glucose-modulating effect especially with gender implication. Methods Twenty-five (25) male and 25 female rats were separately and blindly allocated into five identical groups (n = 5/group). Group I (control) received 0.2 ml/kg distilled water. Groups II and III both received 2.90 mg/kg artesunate on day one, but 1.45 mg/kg from day two till day five and day fifteen respectively. Groups IV and V both received 8.70 mg/kg artesunate on day one, but 4.35 mg/kg artesunate from day two till day five and day fifteen respectively. Results In male rats, glucose was reduced by both doses of artesunate at 5 days but increased by high dose at 15 days. Artesunate increased glycogen concentration at short duration which normalised at long duration in both genders. Artesunate increased G6P concentration only in male rats at 15 days but reduced G6Pase activity in male and female rats (except in those that received low and high doses of artesunate for 15 days). Artesunate increased insulin only in male rats treated with low dose artesunate for 5 days. Artesunate increased cortisol concentration in male but reduced it in female rats. Artesunate decreased glucagon concentration except in female rats treated with high dose for 5 days. Artesunate increased oestrogen concentration in male rats that received low dose artesunate for 5 days but reduced it in female rats that received high dose for 15 days. Conclusions Artesunate reduces plasma glucose by reducing plasma glucagon concentrations and inhibiting liver glycogenolysis via inhibition of G6Pase activity in both sexes. Increase in insulin concentration contributed to the reduction in blood glucose caused by artesunate in male but not female rats; and artesunate-induced increase in G6P, a substrate for G6PD, could enhance NADPH generation and antioxidant enzyme activation in male rats. Reducing glucagon concentration and inhibiting G6Pase activity in both genders. . Increasing plasma insulin in male but not in female. Increasing G6P, a substrate for G6PD, in male rats.
Collapse
Affiliation(s)
- Abdullateef Isiaka Alagbonsi
- Department of Clinical Biology (Physiology unit), School of Medicine and Pharmacy, University of Rwanda College of Medicine and Health Sciences, Huye, Rwanda
| | - Toyin Mohammad Salman
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria
| | - Sheu Oluwadare Sulaiman
- Department of Physiology, Kampala International University Western Campus, Ishaka Bushenyi, Uganda.,Department of Cell Biology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Kafayat Anike Adedini
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria
| | - Susan Kebu
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Kwara, Nigeria
| |
Collapse
|
34
|
8-Week Supplementation of 2S-Hesperidin Modulates Antioxidant and Inflammatory Status after Exercise until Exhaustion in Amateur Cyclists. Antioxidants (Basel) 2021; 10:antiox10030432. [PMID: 33799833 PMCID: PMC8000657 DOI: 10.3390/antiox10030432] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/05/2021] [Accepted: 03/08/2021] [Indexed: 12/30/2022] Open
Abstract
Both acute and chronic ingestion of 2S-hesperidin have shown antioxidant and anti-inflammatory effects in animal studies, but so far, no one has studied this effect of chronic ingestion in humans. The main objective was to evaluate whether an 8-week intake of 2S-hesperidin had the ability to modulate antioxidant-oxidant and inflammatory status in amateur cyclists. A parallel, randomized, double-blind, placebo-controlled trial study was carried out with two groups (500 mg/d 2S-hesperidin; n = 20 and 500 mg/d placebo; n = 20). An incremental test was performed to determine the working zones in a rectangular test, which was used to analyze for changes in antioxidant and inflammatory biomarkers. After 2S-hesperidin ingestion, we found in the rectangular test: (1) an increase in superoxide dismutase (SOD) after the exercise phase until exhaustion (p = 0.045) and the acute recovery phase (p = 0.004), (2) a decrease in the area under the oxidized glutathione curve (GSSG) (p = 0.016), and (3) a decrease in monocyte chemoattractant protein 1 (MCP1) after the acute recovery phase (p = 0.004), post-intervention. Chronic 2S-hesperidin supplementation increased endogenous antioxidant capacity (↑SOD) after maximal effort and decreased oxidative stress (↓AUC-GSSG) during the rectangular test, decreasing inflammation (↓MCP1) after the acute recovery phase.
Collapse
|
35
|
Endothelial response to glucose: dysfunction, metabolism, and transport. Biochem Soc Trans 2021; 49:313-325. [PMID: 33522573 DOI: 10.1042/bst20200611] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/23/2020] [Accepted: 01/04/2021] [Indexed: 02/07/2023]
Abstract
The endothelial cell response to glucose plays an important role in both health and disease. Endothelial glucose-induced dysfunction was first studied in diabetic animal models and in cells cultured in hyperglycemia. Four classical dysfunction pathways were identified, which were later shown to result from the common mechanism of mitochondrial superoxide overproduction. More recently, non-coding RNA, extracellular vesicles, and sodium-glucose cotransporter-2 inhibitors were shown to affect glucose-induced endothelial dysfunction. Endothelial cells also metabolize glucose for their own energetic needs. Research over the past decade highlighted how manipulation of endothelial glycolysis can be used to control angiogenesis and microvascular permeability in diseases such as cancer. Finally, endothelial cells transport glucose to the cells of the blood vessel wall and to the parenchymal tissue. Increasing evidence from the blood-brain barrier and peripheral vasculature suggests that endothelial cells regulate glucose transport through glucose transporters that move glucose from the apical to the basolateral side of the cell. Future studies of endothelial glucose response should begin to integrate dysfunction, metabolism and transport into experimental and computational approaches that also consider endothelial heterogeneity, metabolic diversity, and parenchymal tissue interactions.
Collapse
|
36
|
Identification, Characterization, and Stress Responsiveness of Glucose-6-phosphate Dehydrogenase Genes in Highland Barley. PLANTS 2020; 9:plants9121800. [PMID: 33353078 PMCID: PMC7766724 DOI: 10.3390/plants9121800] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 12/08/2020] [Accepted: 12/13/2020] [Indexed: 01/22/2023]
Abstract
G6PDH provides intermediate metabolites and reducing power (nicotinamide adenine dinucleotide phosphate, NADPH) for plant metabolism, and plays a pivotal role in the cellular redox homeostasis. In this study, we cloned five G6PDH genes (HvG6PDH1 to HvG6PDH5) from highland barley and characterized their encoded proteins. Functional analysis of HvG6PDHs in E. coli showed that HvG6PDH1 to HvG6PDH5 encode the functional G6PDH proteins. Subcellular localization and phylogenetic analysis indicated that HvG6PDH2 and HvG6PDH5 are localized in the cytoplasm, while HvG6PDH1, HvG6PDH3, and HvG6PDH4 are plastidic isoforms. Analysis of enzymatic activities and gene expression showed that HvG6PDH1 to HvG6PDH4 are involved in responses to salt and drought stresses. The cytosolic HvG6PDH2 is the major isoform against oxidative stress. HvG6PDH5 may be a house-keeping gene. In addition, HvG6PDH1 to HvG6PDH4 and their encoded enzymes responded to jasmonic acid (JA) and abscisic acid (ABA) treatments, implying that JA and ABA are probably critical regulators of HvG6PDHs (except for HvG6PDH5). Reactive oxygen species analysis showed that inhibition of cytosolic and plastidic G6PDH activities leads to increased H2O2 and O2− contents in highland barley under salt and drought stresses. These results suggest that G6PDH can maintain cellular redox homeostasis and that cytosolic HvG6PDH2 is an irreplaceable isoform against oxidative stress in highland barley.
Collapse
|
37
|
Ou Z, Chen Y, Li J, Ouyang F, Liu G, Tan S, Huang W, Gong X, Zhang Y, Liang Z, Deng W, Xing S, Zeng J. Glucose-6-phosphate dehydrogenase deficiency and stroke outcomes. Neurology 2020; 95:e1471-e1478. [PMID: 32651291 DOI: 10.1212/wnl.0000000000010245] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Accepted: 03/16/2020] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To assess the risk of glucose-6-phosphate dehydrogenase (G6PD) on stroke prognosis, we compared outcomes between patients with stroke with and without G6PD deficiency. METHODS The study recruited 1,251 patients with acute ischemic stroke. Patients were individually categorized into G6PD-deficiency and non-G6PD-deficiency groups according to G6PD activity upon admission. The primary endpoint was poor outcome at 3 months defined by a modified Rankin Scale (mRS) score ≥2 (including disability and death). Secondary outcomes included the overall mRS score at 3 months and in-hospital death and all death within 3 months. Logistic regression and Cox models, adjusted for potential confounders, were fitted to estimate the association of G6PD deficiency with the outcomes. RESULTS Among 1,251 patients, 150 (12.0%) were G6PD-deficient. Patients with G6PD deficiency had higher proportions of large-artery atherosclerosis (odds ratio [OR] 1.53, 95% confidence interval [CI] 1.09-2.17) and stroke history (OR 1.93, 95% CI 1.26-2.90) compared to the non-G6PD-deficient group. The 2 groups differed significantly in the overall mRS score distribution (adjusted common OR 1.57, 95% CI 1.14-2.17). Patients with G6PD deficiency had higher rates of poor outcome at 3 months (adjusted OR 1.73, 95% CI 1.08-2.76; adjusted absolute risk increase 13.0%, 95% CI 2.4%-23.6%). The hazard ratio of in-hospital death for patients with G6PD-deficiency was 1.46 (95% CI 1.37-1.84). CONCLUSIONS G6PD deficiency is associated with the risk of poor outcome at 3 months after ischemic stroke and may increase the risk of in-hospital death. These findings suggest the rationality of G6PD screening in patients with stroke.
Collapse
Affiliation(s)
- Zilin Ou
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China.
| | - Yicong Chen
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Jianle Li
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Fubing Ouyang
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Gang Liu
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Shuangquan Tan
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Weixian Huang
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Xiao Gong
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Yusheng Zhang
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Zhijian Liang
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Weisheng Deng
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China
| | - Shihui Xing
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China.
| | - Jinsheng Zeng
- From Section II (S.X.), Department of Neurology (Z.O., Y.C., J.L., F.O., G.L., S.T., W.H., J.Z.), The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, Guangzhou; Department of Epidemiology and Biostatistics (X.G.), School of Public Health, Guangdong Pharmaceutical University; Department of Neurology and Stroke Center (Y.Z.), The First Affiliated Hospital of Jinan University, Guangzhou; Department of Neurology (Z.L.), The First Affiliated Hospital of Guangxi Medical University, Nanning; and Department of Neurology (W.D.), Meizhou Hospital Affiliated to Sun Yat-sen University, China.
| |
Collapse
|
38
|
Bhardwaj V, He J. Reactive Oxygen Species, Metabolic Plasticity, and Drug Resistance in Cancer. Int J Mol Sci 2020; 21:ijms21103412. [PMID: 32408513 PMCID: PMC7279373 DOI: 10.3390/ijms21103412] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 05/11/2020] [Indexed: 01/29/2023] Open
Abstract
The metabolic abnormality observed in tumors is characterized by the dependence of cancer cells on glycolysis for their energy requirements. Cancer cells also exhibit a high level of reactive oxygen species (ROS), largely due to the alteration of cellular bioenergetics. A highly coordinated interplay between tumor energetics and ROS generates a powerful phenotype that provides the tumor cells with proliferative, antiapoptotic, and overall aggressive characteristics. In this review article, we summarize the literature on how ROS impacts energy metabolism by regulating key metabolic enzymes and how metabolic pathways e.g., glycolysis, PPP, and the TCA cycle reciprocally affect the generation and maintenance of ROS homeostasis. Lastly, we discuss how metabolic adaptation in cancer influences the tumor’s response to chemotherapeutic drugs. Though attempts of targeting tumor energetics have shown promising preclinical outcomes, the clinical benefits are yet to be fully achieved. A better understanding of the interaction between metabolic abnormalities and involvement of ROS under the chemo-induced stress will help develop new strategies and personalized approaches to improve the therapeutic efficiency in cancer patients.
Collapse
Affiliation(s)
- Vikas Bhardwaj
- College of Pharmacy, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| | - Jun He
- Department of Pathology, Anatomy & Cell Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Correspondence:
| |
Collapse
|
39
|
Panday S, Talreja R, Kavdia M. The role of glutathione and glutathione peroxidase in regulating cellular level of reactive oxygen and nitrogen species. Microvasc Res 2020; 131:104010. [PMID: 32335268 DOI: 10.1016/j.mvr.2020.104010] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/20/2020] [Accepted: 04/20/2020] [Indexed: 12/23/2022]
Abstract
Glutathione (GSH) and GSH/glutathione peroxidase (GPX) enzyme system is essential for normal intracellular homeostasis and gets disturbed under pathophysiologic conditions including endothelial dysfunction. Overproduction of reactive oxidative species (ROS) and reactive nitrogen species (RNS) including superoxide (O2•-), and the loss of nitric oxide (NO) bioavailability is a characteristic of endothelial dysfunction. The GSH/GPX system play an important role in eliminating ROS/RNS. Studies have provided important information regarding the interactions of ROS/RNS with the GSH/GPX in biological systems; however, it is not clear how this cross talk affect these reactive species and GSH/GPX enzyme system, under physiologic and oxidative/nitrosative stress conditions. In the present study, we developed a detailed endothelial cell kinetic model to understand the relationship amongst the key enzyme systems including GSH, GPX, peroxiredoxin (Prx) and reactive species, such as hydrogen peroxide (H2O2), peroxynitrite (ONOO-), and dinitrogen trioxide (N2O3). Our simulation results showed that the alterations in the generation rates of O2•- and NO led to the formation of a wide range of ROS and RNS. Simulations performed by varying the ratio of O2•- to NO generation rates as well as GSH and GPX concentrations showed that the GPX reducing capacity was dependent on GSH availability, level of oxidative/nitrosative stress, and can be attributed to N2O3 levels, but not to H2O2 and ONOO-. Our results showed that N2O3 mediated switch-like depletion in GSH and the incorporation of Prx had no considerable effect on the ROS/RNS species other than ONOO- and H2O2. The analysis presented in this study will improve our understanding of vascular diseases in which the levels and oxidation states of GSH, GPX and/or Prx are significantly altered and pharmacological interventions show limited benefits.
Collapse
Affiliation(s)
- Sheetal Panday
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| | - Raghav Talreja
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America
| | - Mahendra Kavdia
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, United States of America.
| |
Collapse
|
40
|
Tu D, Gao Y, Yang R, Guan T, Hong JS, Gao HM. The pentose phosphate pathway regulates chronic neuroinflammation and dopaminergic neurodegeneration. J Neuroinflammation 2019; 16:255. [PMID: 31805953 PMCID: PMC6896486 DOI: 10.1186/s12974-019-1659-1] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 11/26/2019] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Metabolic dysfunction and neuroinflammation are increasingly implicated in Parkinson's disease (PD). The pentose phosphate pathway (PPP, a metabolic pathway parallel to glycolysis) converts glucose-6-phosphate into pentoses and generates ribose-5-phosphate and NADPH thereby governing anabolic biosynthesis and redox homeostasis. Brains and immune cells display high activity of glucose-6-phosphate dehydrogenase (G6PD), the rate-limiting enzyme of the PPP. A postmortem study reveals dysregulation of G6PD enzyme in brains of PD patients. However, spatial and temporal changes in activity/expression of G6PD in PD remain undetermined. More importantly, it is unclear how dysfunction of G6PD and the PPP affects neuroinflammation and neurodegeneration in PD. METHODS We examined expression/activity of G6PD and its association with microglial activation and dopaminergic neurodegeneration in multiple chronic PD models generated by an intranigral/intraperitoneal injection of LPS, daily subcutaneous injection of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) for 6 days, or transgenic expression of A53T α-synuclein. Primary microglia were transfected with G6PD siRNAs and treated with lipopolysaccharide (LPS) to examine effects of G6PD knockdown on microglial activation and death of co-cultured neurons. LPS alone or with G6PD inhibitor(s) was administrated to mouse substantia nigra or midbrain neuron-glia cultures. While histological and biochemical analyses were conducted to examine microglial activation and dopaminergic neurodegeneration in vitro and in vivo, rotarod behavior test was performed to evaluate locomotor impairment in mice. RESULTS Expression and activity of G6PD were elevated in LPS-treated midbrain neuron-glia cultures (an in vitro PD model) and the substantia nigra of four in vivo PD models. Such elevation was positively associated with microglial activation and dopaminergic neurodegeneration. Furthermore, inhibition of G6PD by 6-aminonicotinamide and dehydroepiandrosterone and knockdown of microglial G6PD attenuated LPS-elicited chronic dopaminergic neurodegeneration. Mechanistically, microglia with elevated G6PD activity/expression produced excessive NADPH and provided abundant substrate to over-activated NADPH oxidase (NOX2) leading to production of excessive reactive oxygen species (ROS). Knockdown and inhibition of G6PD ameliorated LPS-triggered production of ROS and activation of NF-кB thereby dampening microglial activation. CONCLUSIONS Our findings indicated that G6PD-mediated PPP dysfunction and neuroinflammation exacerbated each other mediating chronic dopaminergic neurodegeneration and locomotor impairment. Insight into metabolic-inflammatory interface suggests that G6PD and NOX2 are potential therapeutic targets for PD.
Collapse
Affiliation(s)
- Dezhen Tu
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Yun Gao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Ru Yang
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
| | - Tian Guan
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China
| | - Jau-Shyong Hong
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA
| | - Hui-Ming Gao
- MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Institute for Brain Sciences, Nanjing University, 12 Xuefu Road, Nanjing, 210061, Jiangsu Province, China.
- Neurobiology Laboratory, National Institute of Environmental Health Sciences/National Institutes of Health, Research Triangle Park, Durham, NC, 27709, USA.
| |
Collapse
|
41
|
Yang HC, Wu YH, Yen WC, Liu HY, Hwang TL, Stern A, Chiu DTY. The Redox Role of G6PD in Cell Growth, Cell Death, and Cancer. Cells 2019; 8:cells8091055. [PMID: 31500396 PMCID: PMC6770671 DOI: 10.3390/cells8091055] [Citation(s) in RCA: 141] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Revised: 09/02/2019] [Accepted: 09/07/2019] [Indexed: 02/07/2023] Open
Abstract
The generation of reducing equivalent NADPH via glucose-6-phosphate dehydrogenase (G6PD) is critical for the maintenance of redox homeostasis and reductive biosynthesis in cells. NADPH also plays key roles in cellular processes mediated by redox signaling. Insufficient G6PD activity predisposes cells to growth retardation and demise. Severely lacking G6PD impairs embryonic development and delays organismal growth. Altered G6PD activity is associated with pathophysiology, such as autophagy, insulin resistance, infection, inflammation, as well as diabetes and hypertension. Aberrant activation of G6PD leads to enhanced cell proliferation and adaptation in many types of cancers. The present review aims to update the existing knowledge concerning G6PD and emphasizes how G6PD modulates redox signaling and affects cell survival and demise, particularly in diseases such as cancer. Exploiting G6PD as a potential drug target against cancer is also discussed.
Collapse
Affiliation(s)
- Hung-Chi Yang
- Department of Medical Laboratory Science and Biotechnology, Yuanpei University of Medical Technology, Hsinchu, Taiwan.
| | - Yi-Hsuan Wu
- Research Center for Chinese Herbal Medicine, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| | - Wei-Chen Yen
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Hui-Ya Liu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
| | - Tsong-Long Hwang
- Research Center for Food and Cosmetic Safety, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
- Graduate Institute of Natural Products, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Chinese Herbal Medicine Research Team, Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Anaesthesiology, Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Department of Chemical Engineering, Ming Chi University of Technology, New Taipei City, Taiwan.
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
| | - Arnold Stern
- New York University School of Medicine, New York, NY, USA.
| | - Daniel Tsun-Yee Chiu
- Department of Medical Biotechnology and Laboratory Sciences, College of Medicine, Chang Gung University, Taoyuan, Taiwan.
- Research Center for Chinese Herbal Medicine, Graduate Institute of Health Industry Technology, College of Human Ecology, Chang Gung University of Science and Technology, Taoyuan, Taiwan.
- Department of Pediatric Hematology/Oncology, Linkou Chang Gung Memorial Hospital, Taoyuan, Taiwan.
- Healthy Aging Research Center, Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
42
|
Li X, Sun X, Carmeliet P. Hallmarks of Endothelial Cell Metabolism in Health and Disease. Cell Metab 2019; 30:414-433. [PMID: 31484054 DOI: 10.1016/j.cmet.2019.08.011] [Citation(s) in RCA: 278] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 08/10/2019] [Accepted: 08/12/2019] [Indexed: 01/13/2023]
Abstract
In 2009, it was postulated that endothelial cells (ECs) would only be able to execute the orders of growth factors if these cells would accordingly adapt their metabolism. Ten years later, it has become clear that ECs, often differently from other cell types, rely on distinct metabolic pathways to survive and form new blood vessels; that manipulation of EC metabolic pathways alone (even without changing angiogenic signaling) suffices to alter vessel sprouting; and that perturbations of these metabolic pathways can underlie excess formation of new blood vessels (angiogenesis) in cancer and ocular diseases. Initial proof of evidence has been provided that targeting (normalizing) these metabolic perturbations in diseased ECs and delivery of metabolites deserve increasing attention as novel therapeutic approaches for inhibiting or stimulating vessel growth in multiple disorders.
Collapse
Affiliation(s)
- Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China.
| | - Xiaodong Sun
- Department of Ophthalmology, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, P.R. China
| | - Peter Carmeliet
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven B-3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven B-3000, Belgium.
| |
Collapse
|
43
|
Brothers RM, Fadel PJ, Keller DM. Racial disparities in cardiovascular disease risk: mechanisms of vascular dysfunction. Am J Physiol Heart Circ Physiol 2019; 317:H777-H789. [PMID: 31397168 DOI: 10.1152/ajpheart.00126.2019] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease (CVD) accounts for a third of all deaths in the United States making it the leading cause of morbidity and mortality. Although CVD affects individuals of all races/ethnicities, the prevalence of CVD is highest in non-Hispanic black (BL) individuals relative to other populations. The mechanism(s) responsible for elevated CVD risk in the BL population remains incompletely understood. However, impaired vascular vasodilator capacity and exaggerated vascular vasoconstrictor responsiveness are likely contributing factors, both of which are present even in young, otherwise healthy BL individuals. Within this review, we highlight some historical and recent data, collected from our laboratories, of impaired vascular function, in terms of reduced vasodilator capacity and heightened vasoconstrictor responsiveness, in the peripheral and cerebral circulations in BL individuals. We provide data that such impairments may be related to elevated oxidative stress and subsequent reduction in nitric oxide bioavailability. In addition, divergent mechanisms of impaired vasodilatory capacity between BL men and women are discussed. Finally, we propose several directions where future research is needed to fill in knowledge gaps, which will allow for better understanding of the mechanisms contributing to impaired vascular function in this population. Ultimately, this information will allow for better lifestyle and therapeutic approaches to be implemented in an effort to minimize the increased CVD burden in the BL population.
Collapse
Affiliation(s)
- R Matthew Brothers
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - Paul J Fadel
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| | - David M Keller
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
44
|
Elarabany N, Bahnasawy M. Comparative and Interactive Biochemical Effects of Sub-Lethal Concentrations of Cadmium and Lead on Some Tissues of the African Catfish ( Clarias gariepinus). Toxicol Res 2019; 35:249-255. [PMID: 31341554 PMCID: PMC6629445 DOI: 10.5487/tr.2019.35.3.249] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 11/15/2018] [Accepted: 11/20/2018] [Indexed: 11/22/2022] Open
Abstract
Cadmium is a strong toxic heavy metal which presents in paints and liquid wastes and causes oxidative stress in fish. On the other hand, lead is widely used for different purposes, e.g. lead pipes, it targets vital organs such as liver and kidney causing biochemical alterations. The present study evaluates the effects of 60 days exposure to Cd and Pb either single or combined together in African catfish. Sixty-four fishes were divided into 3 groups and exposed to CdCl2 (7.02 mg/L) or PbCl2 (69.3 mg/L) or a combination of them along with control group. Activities of acid phosphatase (ACP), lactate dehydrogenase (LDH) and glucose-6-phosphate dehydrogenase (G-6-PDH) were estimated. Moreover, gill, liver and kidney were assayed for activities of superoxide dismutase (SOD), catalase (CAT) and levels of glutathione (GSH) and malondialdehyde (MDA). Individual exposure showed that both Cd and Pb significantly decreased LDH activity and SOD activity in the kidney. Pb significantly increased G-6-PDH activity and decreased GSH level in the gill. CAT activity in liver and kidney elevated significantly on Cd exposure while lead caused a significant depletion in the liver and significant elevation in the kidney. Both Cd and Pb significantly increased MDA levels in liver and kidney while Pb increased its level in gills. The combined exposure resulted in normalization of LDH, G-6-PDH activity, and CAT activity in liver and kidney as well as GSH level in both tissues and MDA in gill and kidney. The combination increased SOD activity and MDA level in liver and decreased SOD activity in kidney and GSH level in gills. In conclusion, the antioxidant system of African catfish was adversely affected by prolonged exposure to Cd and Pb. The combined exposure caused less damage than individual exposure and returned most parameters to those of controls.
Collapse
Affiliation(s)
- Naglaa Elarabany
- Zoology Department, Faculty of Science, Damietta University, Damietta, Egypt
| | - Mohammed Bahnasawy
- Zoology Department, Faculty of Science, Damietta University, Damietta, Egypt
| |
Collapse
|
45
|
Tumova S, Kerimi A, Williamson G. Long term treatment with quercetin in contrast to the sulfate and glucuronide conjugates affects HIF1α stability and Nrf2 signaling in endothelial cells and leads to changes in glucose metabolism. Free Radic Biol Med 2019; 137:158-168. [PMID: 31029788 DOI: 10.1016/j.freeradbiomed.2019.04.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 04/17/2019] [Accepted: 04/18/2019] [Indexed: 01/08/2023]
Abstract
Endothelial functionality profoundly contributes to cardiovascular health. The effects of flavonoids shown to improve endothelial performance include regulating blood pressure by modulating endothelial nitric oxide synthase and NADPH oxidases, but their impact on glucose uptake and metabolism has not been explored. We treated human umbilical vein endothelial cells (HUVEC) with the flavonoid quercetin and its circulating metabolites acutely and chronically, then assessed glucose uptake, glucose metabolism, gene transcription and protein expression. Acute treatment had no effect on glucose uptake, ruling out any direct interaction with sugar transporters. Long term treatment with quercetin, but not quercetin 3-O-glucuronide or 3'-O-sulfate, significantly increased glucose uptake. Heme oxygenase-1 (HO-1) was induced by quercetin but not its conjugates, but was not implicated in the glucose uptake stimulation since hemin, a classical inducer of HO-1, did not affect glucose metabolism. Quercetin increased stability of the transcription factor hypoxia induced factor 1α (HIF1α), a powerful stimulant of glucose metabolism, which was also paralleled by treatment with a prolyl-4-hydroxylase inhibitor dimethyloxalylglycine (DMOG). 6-Phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), which regulates the rate of glycolysis, was upregulated by both quercetin and DMOG. Pyruvate dehydrogenase kinase (PDK) isoforms regulate pyruvate dehydrogenase; PDK2 and PDK4 were down-regulated by both effectors, but only DMOG also upregulated PDK1 and PDK3. Quercetin, but not DMOG, increased glucose-6-phosphate dehydrogenase. Chronic quercetin treatment also stimulated glucose transport across the HUVEC monolyer in a 3D culture model. Gene expression of several flavonoid transporters was repressed by quercetin, but this was either abolished (Organic anion transporter polypeptide 4C1) or reversed (Multidrug resistance gene 1) by both conjugates. We conclude that quercetin and its circulating metabolites differentially modulate glucose uptake/metabolism in endothelial cells, through effects on HIF1α and transcriptional regulation of energy metabolism.
Collapse
Affiliation(s)
- Sarka Tumova
- School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT, UK
| | - Asimina Kerimi
- School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT, UK
| | - Gary Williamson
- School of Food Science and Nutrition, University of Leeds, Leeds, LS2 9JT, UK; Department of Nutrition, Dietetics and Food, School of Clinical Sciences at Monash Health, Faculty of Medicine, Nursing and Health Sciences, Monash University, Notting Hill BASE Facility, 264 Ferntree Gully Road, Notting Hill, VIC, 3168, Australia.
| |
Collapse
|
46
|
Li Y, Wan YY, Zhu B. Immune Cell Metabolism in Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1011:163-196. [PMID: 28875490 DOI: 10.1007/978-94-024-1170-6_5] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Tumor microenvironment (TME) is composed of tumor cells, immune cells, cytokines, extracellular matrix, etc. The immune system and the metabolisms of glucose, lipids, amino acids, and nucleotides are integrated in the tumorigenesis and development. Cancer cells and immune cells show metabolic reprogramming in the TME, which intimately links immune cell functions and edits tumor immunology. Recent findings in immune cell metabolism hold the promising possibilities toward clinical therapeutics for treating cancer. This chapter introduces the updated understandings of metabolic reprogramming of immune cells in the TME and suggests new directions in manipulation of immune responses for cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Yongsheng Li
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Yisong Y Wan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China.
| |
Collapse
|
47
|
Soliman HAM, Hamed M, Lee JS, Sayed AEDH. Protective effects of a novel pyrazolecarboxamide derivative against lead nitrate induced oxidative stress and DNA damage in Clarias gariepinus. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 247:678-684. [PMID: 30711823 DOI: 10.1016/j.envpol.2019.01.074] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 01/15/2019] [Accepted: 01/20/2019] [Indexed: 02/07/2023]
Abstract
Pyrazole derivatives display diverse biological and pharmacological activities. The aim of this study is to investigate the antioxidant properties of a novel pyrazolecarboxamide derivative (4-amino-N-[(4-chlorophenyl)]-3-methyl-1-phenyl-1H-thieno [2, 3-c] pyrazole-5-carboxamide) in African catfish, Clarias gariepinus, exposed to 1 mg/L PbNO3. Fish were intramuscularly injected with pyrazole-5-carboxamidederivative according to the following groupings: Group 1 (control), Group 2 (1 mg/L lead nitrate), Group 3 (1 mg/L lead nitrate + 5 mg pyrazole derivative/kg body weight), and Group 4 (1 mg/L lead nitrate + 10 mg pyrazole derivative/kg body weight) for two weeks and four weeks. Lead nitrate (1 mg/L) caused significant elevation of serum aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, uric acid, cholesterol, and glucose-6-phosphate dehydrogenase (G6PDH) compared to the control group after two and four weeks of exposure, while serum total lipids, alkaline phosphatase (ALP), and lactate dehydrogenase (LDH) were significantly reduced compared to the control group. Furthermore, levels of antioxidant enzymes superoxide dismutase (SOD) and catalase (CAT) and total antioxidant capacity (TAC) were reduced in group 2 compared to the control group. However, in group 2, hepatic lipid peroxidation (LPO) and DNA fragmentation percentage were significantly increased compared to the control group. Histopathological changes in the liver of lead-exposed groups included marked disturbance of hepatic tissue organization, degeneration of hepatocytes, dilation of blood sinusoids and the central vein as well as necrosis. Injection of pyrazole derivative for two weeks and four weeks reversed alterations in biochemical parameters, antioxidant biomarkers, lipid peroxidation, hepatic DNA damage, and histopathological changes in liver tissue induced by 1 mg/L lead nitrate. This amelioration was higher in response to high-dose pyrazole derivative (10 mg) at the fourth week of exposure, showing concentration-and time-dependency. Overall, the sensitized derivative pyrazolecarboxamide is likely a useful tool to minimize the effects of lead toxicity due to its potent antioxidant activity.
Collapse
Affiliation(s)
- Hamdy A M Soliman
- Department of Zoology, Faculty of Science, Sohag University, 8562, Sohag, Egypt
| | - Mohamed Hamed
- Department of Zoology, Faculty of Science, Al Azhar University (Assiut Branch), 71524, Assiut, Egypt
| | - Jae-Seong Lee
- Department of Biological Science, College of Science, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Alaa El-Din H Sayed
- Department of Zoology, Faculty of Science, Assiut University, 71516, Assiut, Egypt.
| |
Collapse
|
48
|
Teuwen LA, Geldhof V, Carmeliet P. How glucose, glutamine and fatty acid metabolism shape blood and lymph vessel development. Dev Biol 2019; 447:90-102. [DOI: 10.1016/j.ydbio.2017.12.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 10/26/2017] [Accepted: 12/01/2017] [Indexed: 12/18/2022]
|
49
|
Abstract
SIGNIFICANCE Angiogenesis is the formation of new vessels that sprout from existing vessels. This process is highly complex and requires a coordinated shift of the endothelial phenotype from a quiescent cell in the vessel wall into a migrating or proliferating cell. Such change in the life of the endothelial cell is induced by a variety of factors such as hypoxia, metabolic changes, or cytokines. Recent Advances: Within the last years, it became clear that the cellular redox state and oxidation of signaling molecules or phosphatases are critical modulators in angiogenesis. CRITICAL ISSUES According to the wide variety of stimuli that induce angiogenesis, a complex signaling network is needed to support a coordinated response of the endothelial cell. Reactive oxygen species (ROS) now are second messengers that either directly oxidize a target molecule or initiate a cascade of redox sensitive steps that transmit the signal. Further Directions: For the understanding of redox signaling, it is essential to recognize and accept that ROS do not represent master regulators of angiogenetic processes. They rather modulate existing signal cascades. This review summarizes some current findings on redox signaling in angiogenesis.
Collapse
Affiliation(s)
- Katrin Schröder
- 1 Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany.,2 German Center for Cardiovascular Research (DZHK), Rhine-Main, Frankfurt, Germany
| |
Collapse
|
50
|
Wang M, Hu J, Yan L, Yang Y, He M, Wu M, Li Q, Gong W, Yang Y, Wang Y, Handy DE, Lu B, Hao C, Wang Q, Li Y, Hu R, Stanton RC, Zhang Z. High glucose-induced ubiquitination of G6PD leads to the injury of podocytes. FASEB J 2019; 33:6296-6310. [PMID: 30785802 DOI: 10.1096/fj.201801921r] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Oxidative stress contributes substantially to podocyte injury, which plays an important role in the development of diabetic kidney disease. The mechanism of hyperglycemia-induced oxidative stress in podocytes is not fully understood. Glucose-6-phosphate dehydrogenase (G6PD) is critical in maintaining NADPH, which is an important cofactor for the antioxidant system. Here, we hypothesized that high glucose induced ubiquitination and degradation of G6PD, which injured podocytes by reactive oxygen species (ROS) accumulation. We found that G6PD protein expression was decreased in kidneys of both diabetic patients and diabetic rodents. G6PD activity was also reduced in diabetic mice. Overexpressing G6PD reversed redox imbalance and podocyte apoptosis induced by high glucose and palmitate. Inhibition of G6PD with small interfering RNA induced podocyte apoptosis. In kidneys of G6PD-deficient mice, podocyte apoptosis was significantly increased. Interestingly, high glucose had no effect on G6PD mRNA expression. Decreased G6PD protein expression was mediated by the ubiquitin proteasome pathway. We found that the von Hippel-Lindau (VHL) protein, an E3 ubiquitin ligase subunit, directly bound to G6PD and degraded G6PD through ubiquitylating G6PD on K366 and K403. In summary, our data suggest that high glucose induces ubiquitination of G6PD by VHL E3 ubiquitin ligase, which leads to ROS accumulation and podocyte injury.-Wang, M., Hu, J., Yan, L., Yang, Y., He, M., Wu, M., Li, Q., Gong, W., Yang, Y., Wang, Y., Handy, D. E., Lu, B., Hao, C., Wang, Q., Li, Y., Hu, R., Stanton, R. C., Zhang, Z. High glucose-induced ubiquitination of G6PD leads to the injury of podocytes.
Collapse
Affiliation(s)
- Meng Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Ji Hu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Linling Yan
- Department of Endocrinology, The First People's Hospital of Taicang, Suzhou, China
| | - Yeping Yang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Min He
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Meng Wu
- Department of Endocrinology, The Second Affiliated Hospital, Soochow University, Suzhou, China
| | - Qin Li
- Department of Endocrinology and Metabolism, Shanghai Ninth People's Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Wei Gong
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Yang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Yi Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Diane E Handy
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Bin Lu
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Chuanming Hao
- Division of Nephrology, Huashan Hospital, Fudan University, Shanghai, China
| | - Qinghua Wang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China.,Division of Endocrinology and Metabolism, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Yiming Li
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| | - Ronggui Hu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Shanghai, China
| | - Robert C Stanton
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhaoyun Zhang
- Division of Endocrinology and Metabolism, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|