1
|
Keeney MT, Rocha EM, Hoffman EK, Farmer K, Di Maio R, Weir J, Wagner WG, Hu X, Clark CL, Castro SL, Scheirer A, Fazzari M, De Miranda BR, Pintchovski SA, Shrader WD, Pagano PJ, Hastings TG, Greenamyre JT. LRRK2 regulates production of reactive oxygen species in cell and animal models of Parkinson's disease. Sci Transl Med 2024; 16:eadl3438. [PMID: 39356746 DOI: 10.1126/scitranslmed.adl3438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 04/30/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
Oxidative stress has long been implicated in Parkinson's disease (PD) pathogenesis, although the sources and regulation of reactive oxygen species (ROS) production are poorly defined. Pathogenic mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are associated with increased kinase activity and a greater risk of PD. The substrates and downstream consequences of elevated LRRK2 kinase activity are still being elucidated, but overexpression of mutant LRRK2 has been associated with oxidative stress, and antioxidants reportedly mitigate LRRK2 toxicity. Here, using CRISPR-Cas9 gene-edited HEK293 cells, RAW264.7 macrophages, rat primary ventral midbrain cultures, and PD patient-derived lymphoblastoid cells, we found that elevated LRRK2 kinase activity was associated with increased ROS production and lipid peroxidation and that this was blocked by inhibitors of either LRRK2 kinase or NADPH oxidase 2 (NOX2). Oxidative stress induced by the pesticide rotenone was ameliorated by LRRK2 kinase inhibition and was absent in cells devoid of LRRK2. In a rat model of PD induced by rotenone, a LRRK2 kinase inhibitor prevented the lipid peroxidation and NOX2 activation normally seen in nigral dopaminergic neurons in this model. Mechanistically, LRRK2 kinase activity was shown to regulate phosphorylation of serine-345 in the p47phox subunit of NOX2. This, in turn, led to translocation of p47phox from the cytosol to the membrane-associated gp91phox (NOX2) subunit, activation of the NOX2 enzyme complex, and production of ROS. Thus, LRRK2 kinase activity may drive cellular ROS production in PD through the regulation of NOX2 activity.
Collapse
Affiliation(s)
- Matthew T Keeney
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Emily M Rocha
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Eric K Hoffman
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kyle Farmer
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roberto Di Maio
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Julie Weir
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Weston G Wagner
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Xiaoping Hu
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Courtney L Clark
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Sandra L Castro
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Abigail Scheirer
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Briana R De Miranda
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | - Patrick J Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Teresa G Hastings
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Blaise O, Duchesne C, Capuzzo E, Nahori MA, Fernandes J, Connor MG, Hamon MA, Pizarro-Cerda J, Lataillade JJ, McGuckin C, Rousseau A, Banzet S, Dussurget O, Frescaline N. Infected wound repair correlates with collagen I induction and NOX2 activation by cold atmospheric plasma. NPJ Regen Med 2024; 9:28. [PMID: 39358383 PMCID: PMC11447178 DOI: 10.1038/s41536-024-00372-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Cold atmospheric plasma (CAP) is a promising complement to tissue repair and regenerative medicine approaches. CAP has therapeutic potential in infected cutaneous wounds by mechanisms which remain enigmatic. Here, CAP is shown to activate phagocyte NADPH oxidase complex NOX2. CAP induced increased intracellular reactive oxygen species, alleviated by NOX2 inhibitors. Genetic and pharmacological inhibitions of NOX2 in macrophages and bioengineered skin infected with Staphylococcus aureus and treated with CAP reduced intracellular oxidants and increased bacterial survival. CAP triggered Rac activation and phosphorylation of p40phox and p47phox required for NOX2 assembly and activity. Furthermore, CAP induced collagen I expression by fibroblasts. Infection and healing kinetics showed that murine skin wounds infected with S. aureus and treated with CAP are characterized by decreased bacterial burden, increased length of neoepidermis and extracellular matrix formation. Collectively, our findings identify mechanisms triggered by CAP that subdue infection and result in enhanced repair following skin injury.
Collapse
Affiliation(s)
- Océane Blaise
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
- École Polytechnique, Sorbonne Université, CNRS UMR7648, Laboratoire de Physique des Plasmas, Palaiseau, France
| | - Constance Duchesne
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
- École Polytechnique, Sorbonne Université, CNRS UMR7648, Laboratoire de Physique des Plasmas, Palaiseau, France
| | - Elena Capuzzo
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
| | - Marie-Anne Nahori
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité des Toxines Bactériennes, Paris, France
| | | | - Michael G Connor
- Institut Pasteur, Université Paris Cité, Unité Chromatine et Infection, Paris, France
| | - Mélanie A Hamon
- Institut Pasteur, Université Paris Cité, Unité Chromatine et Infection, Paris, France
| | - Javier Pizarro-Cerda
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France
| | | | | | - Antoine Rousseau
- École Polytechnique, Sorbonne Université, CNRS UMR7648, Laboratoire de Physique des Plasmas, Palaiseau, France
| | - Sébastien Banzet
- Centre de Transfusion Sanguine des Armées, Clamart, France
- Institut de Recherche Biomédicale des Armées, INSERM UMRS-MD 1197, Brétigny-sur-Orge, France
| | - Olivier Dussurget
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France.
| | - Nadira Frescaline
- Institut Pasteur, Université Paris Cité, CNRS UMR6047, Unité de Recherche Yersinia, Paris, France.
- Centre de Transfusion Sanguine des Armées, Clamart, France.
| |
Collapse
|
3
|
Totoń-Żurańska J, Mikolajczyk TP, Saju B, Guzik TJ. Vascular remodelling in cardiovascular diseases: hypertension, oxidation, and inflammation. Clin Sci (Lond) 2024; 138:817-850. [PMID: 38920058 DOI: 10.1042/cs20220797] [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: 09/26/2023] [Revised: 06/08/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024]
Abstract
Optimal vascular structure and function are essential for maintaining the physiological functions of the cardiovascular system. Vascular remodelling involves changes in vessel structure, including its size, shape, cellular and molecular composition. These changes result from multiple risk factors and may be compensatory adaptations to sustain blood vessel function. They occur in diverse cardiovascular pathologies, from hypertension to heart failure and atherosclerosis. Dynamic changes in the endothelium, fibroblasts, smooth muscle cells, pericytes or other vascular wall cells underlie remodelling. In addition, immune cells, including macrophages and lymphocytes, may infiltrate vessels and initiate inflammatory signalling. They contribute to a dynamic interplay between cell proliferation, apoptosis, migration, inflammation, and extracellular matrix reorganisation, all critical mechanisms of vascular remodelling. Molecular pathways underlying these processes include growth factors (e.g., vascular endothelial growth factor and platelet-derived growth factor), inflammatory cytokines (e.g., interleukin-1β and tumour necrosis factor-α), reactive oxygen species, and signalling pathways, such as Rho/ROCK, MAPK, and TGF-β/Smad, related to nitric oxide and superoxide biology. MicroRNAs and long noncoding RNAs are crucial epigenetic regulators of gene expression in vascular remodelling. We evaluate these pathways for potential therapeutic targeting from a clinical translational perspective. In summary, vascular remodelling, a coordinated modification of vascular structure and function, is crucial in cardiovascular disease pathology.
Collapse
Affiliation(s)
- Justyna Totoń-Żurańska
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz P Mikolajczyk
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Blessy Saju
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| | - Tomasz J Guzik
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- Department of Internal Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
4
|
Won SJ, Zhang Y, Butler NJ, Kim K, Mocanu E, Nzoutchoum OT, Lakkaraju R, Davis J, Ghosh S, Swanson RA. Stress hyperglycemia exacerbates inflammatory brain injury after stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.594195. [PMID: 38798486 PMCID: PMC11118312 DOI: 10.1101/2024.05.14.594195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Post-stroke hyperglycemia occurs in 30% - 60% of ischemic stroke patients as part of the systemic stress response, but neither clinical evidence nor pre-clinical studies indicate whether post-stroke hyperglycemia affects stroke outcome. Here we investigated this issue using a mouse model of permanent ischemia. Mice were maintained either normoglycemic or hyperglycemic during the interval of 17 - 48 hours after ischemia onset. Post-stroke hyperglycemia was found to increase infarct volume, blood-brain barrier disruption, and hemorrhage formation, and to impair motor recovery. Post-stroke hyperglycemia also increased superoxide formation by peri-infarct microglia/macrophages. In contrast, post-stroke hyperglycemia did not increase superoxide formation or exacerbate motor impairment in p47 phox-/- mice, which cannot form an active superoxide-producing NADPH oxidase-2 complex. These results suggest that hyperglycemia occurring hours-to-days after ischemia can increase oxidative stress in peri-infarct tissues by fueling NADPH oxidase activity in reactive microglia/macrophages, and by this mechanism contribute to worsened functional outcome.
Collapse
|
5
|
Shi X, Li P, Herb M, Liu H, Wang M, Wang X, Feng Y, van Beers T, Xia N, Li H, Prokosch V. Pathological high intraocular pressure induces glial cell reactive proliferation contributing to neuroinflammation of the blood-retinal barrier via the NOX2/ET-1 axis-controlled ERK1/2 pathway. J Neuroinflammation 2024; 21:105. [PMID: 38649885 PMCID: PMC11034147 DOI: 10.1186/s12974-024-03075-x] [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: 01/02/2024] [Accepted: 03/26/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND NADPH oxidase (NOX), a primary source of endothelial reactive oxygen species (ROS), is considered a key event in disrupting the integrity of the blood-retinal barrier. Abnormalities in neurovascular-coupled immune signaling herald the loss of ganglion cells in glaucoma. Persistent microglia-driven inflammation and cellular innate immune system dysregulation often lead to deteriorating retinal degeneration. However, the crosstalk between NOX and the retinal immune environment remains unresolved. Here, we investigate the interaction between oxidative stress and neuroinflammation in glaucoma by genetic defects of NOX2 or its regulation via gp91ds-tat. METHODS Ex vivo cultures of retinal explants from wildtype C57BL/6J and Nox2 -/- mice were subjected to normal and high hydrostatic pressure (Pressure 60 mmHg) for 24 h. In vivo, high intraocular pressure (H-IOP) was induced in C57BL/6J mice for two weeks. Both Pressure 60 mmHg retinas and H-IOP mice were treated with either gp91ds-tat (a NOX2-specific inhibitor). Proteomic analysis was performed on control, H-IOP, and treatment with gp91ds-tat retinas to identify differentially expressed proteins (DEPs). The study also evaluated various glaucoma phenotypes, including IOP, retinal ganglion cell (RGC) functionality, and optic nerve (ON) degeneration. The superoxide (O2-) levels assay, blood-retinal barrier degradation, gliosis, neuroinflammation, enzyme-linked immunosorbent assay (ELISA), western blotting, and quantitative PCR were performed in this study. RESULTS We found that NOX2-specific deletion or activity inhibition effectively attenuated retinal oxidative stress, immune dysregulation, the internal blood-retinal barrier (iBRB) injury, neurovascular unit (NVU) dysfunction, RGC loss, and ON axonal degeneration following H-IOP. Mechanistically, we unveiled for the first time that NOX2-dependent ROS-driven pro-inflammatory signaling, where NOX2/ROS induces endothelium-derived endothelin-1 (ET-1) overexpression, which activates the ERK1/2 signaling pathway and mediates the shift of microglia activation to a pro-inflammatory M1 phenotype, thereby triggering a neuroinflammatory outburst. CONCLUSIONS Collectively, we demonstrate for the first time that NOX2 deletion or gp91ds-tat inhibition attenuates iBRB injury and NVU dysfunction to rescue glaucomatous RGC loss and ON axon degeneration, which is associated with inhibition of the ET-1/ERK1/2-transduced shift of microglial cell activation toward a pro-inflammatory M1 phenotype, highlighting NOX2 as a potential target for novel neuroprotective therapies in glaucoma management.
Collapse
Affiliation(s)
- Xin Shi
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Panpan Li
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital of Cologne, University of Cologne, Goldenfelsstr. 19-21, 50935, Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Hanhan Liu
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Maoren Wang
- Department of Ophthalmology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, 100020, P. R. China
| | - Xiaosha Wang
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Yuan Feng
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany
| | - Tim van Beers
- Institut I für Anatomie, Universitätsklinikum Köln (AöR), Cologne, Germany
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
| | - Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstr. 1, 55131, Mainz, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Rhine-Main, 55131, Mainz, Germany
| | - Verena Prokosch
- Department of Ophthalmology, Faculty of Medicine, University Hospital of Cologne, University of Cologne, 50937, Cologne, Germany.
| |
Collapse
|
6
|
Amadio P, Sandrini L, Zarà M, Barbieri SS, Ieraci A. NADPH-oxidases as potential pharmacological targets for thrombosis and depression comorbidity. Redox Biol 2024; 70:103060. [PMID: 38310682 PMCID: PMC10848036 DOI: 10.1016/j.redox.2024.103060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/06/2024] Open
Abstract
There is a complex interrelationship between the nervous system and the cardiovascular system. Comorbidities of cardiovascular diseases (CVD) with mental disorders, and vice versa, are prevalent. Adults with mental disorders such as anxiety and depression have a higher risk of developing CVD, and people with CVD have an increased risk of being diagnosed with mental disorders. Oxidative stress is one of the many pathways associated with the pathophysiology of brain and cardiovascular disease. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is one of the major generators of reactive oxygen species (ROS) in mammalian cells, as it is the enzyme that specifically produces superoxide. This review summarizes recent findings on the consequences of NOX activation in thrombosis and depression. It also discusses the therapeutic effects and pharmacological strategies of NOX inhibitors in CVD and brain disorders. A better comprehension of these processes could facilitate the development of new therapeutic approaches for the prevention and treatment of the comorbidity of thrombosis and depression.
Collapse
Affiliation(s)
- Patrizia Amadio
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Leonardo Sandrini
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Marta Zarà
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy
| | - Silvia S Barbieri
- Unit of Brain-Heart Axis: Cellular and Molecular Mechanisms, Centro Cardiologico Monzino IRCCS, 20138, Milan, Italy.
| | - Alessandro Ieraci
- Department of Theoretical and Applied Sciences, eCampus University, 22060, Novedrate (CO), Italy; Department of Neuroscience, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156, Milan, Italy.
| |
Collapse
|
7
|
Liu B, Xia L, Li Y, Jiang S, Yu W, Zhang L, Shao XM, Xu Z, Xiao D. Prenatal Nicotine Exposure Raises Male Blood Pressure via FTO-Mediated NOX2/ROS Signaling. Hypertension 2024; 81:240-251. [PMID: 37795601 PMCID: PMC10873091 DOI: 10.1161/hypertensionaha.123.21766] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/16/2023] [Indexed: 10/06/2023]
Abstract
BACKGROUND Cigarette smoking/nicotine exposure in pregnancy shows an increased risk of hypertension in offspring, but the mechanisms are unclear. This study tested the hypothesis that m6A RNA hypomethylation epigenetically regulates vascular NOX (NADPH oxidase) and reactive oxygen species production, contributing to the fetal programming of a hypertensive phenotype in nicotine-exposed offspring. METHODS Pregnant rats were exposed to episodic chronic intermittent nicotine aerosol (CINA) or saline aerosol control from gestational day 4 to day 21, and experiments were performed in 6-month-old adult offspring. RESULTS Antenatal CINA exposure augmented Ang II (angiotensin II)-stimulated blood pressure response in male, but not female offspring. Moreover, CINA increased vascular NOX2 expression and superoxide production exclusively in male offspring. Inhibition of NOX2 with gp91ds-tat, both ex vivo and in vivo, mitigated the CINA-induced elevation in superoxide production and blood pressure response. Notably, CINA enhanced the expression of vascular m6A demethylase FTO (fat mass and obesity-associated protein), while reducing the total vascular m6A abundance and specific m6A methylation of the NOX2 gene. Additionally, ex vivo inhibition of FTO with FB23-2 attenuated CINA-induced increases in vascular NOX2 expression. In vitro experiments using human umbilical vein endothelial cells demonstrated that nicotine dose-dependently upregulated FTO and NOX2 protein abundance, which were reversed by treatment with the FTO inhibitor FB23-2 or FTO knockdown using siRNAs. CONCLUSIONS This study uncovers a new mechanism: m6A demethylase FTO-mediated epigenetic upregulation of vascular NOX2 signaling in CINA-induced hypertensive phenotype. This insight could lead to a therapeutic target for preventing and treating developmental hypertension programming.
Collapse
Affiliation(s)
- Bailin Liu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA 92354
- Perinatology Laboratory, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi 214002, China
| | - Liang Xia
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA 92354
- Department of Surgical Intensive Care Unit, The First Affiliated hospital of Chongqing Medical University, Chongqing 400016, China
| | - Yong Li
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA 92354
| | - Siyi Jiang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA 92354
| | - Wansu Yu
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA 92354
| | - Lubo Zhang
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA 92354
| | - Xuesi M Shao
- Department of Neurobiology, David Geffen School of Medicine at UCLA, University of California at Los Angeles, Los Angeles, CA90095, USA
| | - Zhice Xu
- Perinatology Laboratory, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi 214002, China
- Institute for Fetology, First Hospital of Soochow University, 708 Renmin Road, Suzhou 215006, China
| | - Daliao Xiao
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California, USA 92354
| |
Collapse
|
8
|
Reis J, Gorgulla C, Massari M, Marchese S, Valente S, Noce B, Basile L, Törner R, Cox H, Viennet T, Yang MH, Ronan MM, Rees MG, Roth JA, Capasso L, Nebbioso A, Altucci L, Mai A, Arthanari H, Mattevi A. Targeting ROS production through inhibition of NADPH oxidases. Nat Chem Biol 2023; 19:1540-1550. [PMID: 37884805 DOI: 10.1038/s41589-023-01457-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023]
Abstract
NADPH oxidases (NOXs) are transmembrane enzymes that are devoted to the production of reactive oxygen species (ROS). In cancers, dysregulation of NOX enzymes affects ROS production, leading to redox unbalance and tumor progression. Consequently, NOXs are a drug target for cancer therapeutics, although current therapies have off-target effects: there is a need for isoenzyme-selective inhibitors. Here, we describe fully validated human NOX inhibitors, obtained from an in silico screen, targeting the active site of Cylindrospermum stagnale NOX5 (csNOX5). The hits are validated by in vitro and in cellulo enzymatic and binding assays, and their binding modes to the dehydrogenase domain of csNOX5 studied via high-resolution crystal structures. A high-throughput screen in a panel of cancer cells shows activity in selected cancer cell lines and synergistic effects with KRAS modulators. Our work lays the foundation for the development of inhibitor-based methods for controlling the tightly regulated and highly localized ROS sources.
Collapse
Affiliation(s)
- Joana Reis
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Christoph Gorgulla
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Physics, Faculty of Arts and Sciences, Harvard University, Cambridge, MA, USA
| | - Marta Massari
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Sara Marchese
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Sergio Valente
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Beatrice Noce
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy
| | - Lorenzo Basile
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy
| | - Ricarda Törner
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Huel Cox
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thibault Viennet
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Moon Hee Yang
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | | | | | | | - Lucia Capasso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Antonello Mai
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, Rome, Italy.
| | - Haribabu Arthanari
- Department of Biological Chemistry and Molecular Pharmacology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA.
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - Andrea Mattevi
- Department of Biology and Biotechnology Lazzaro Spallanzani, University of Pavia, Pavia, Italy.
| |
Collapse
|
9
|
Ofori K, Ghosh A, Verma DK, Wheeler D, Cabrera G, Seo JB, Kim YH. A Novel NOX Inhibitor Alleviates Parkinson's Disease Pathology in PFF-Injected Mice. Int J Mol Sci 2023; 24:14278. [PMID: 37762579 PMCID: PMC10531511 DOI: 10.3390/ijms241814278] [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: 08/15/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Oxidative stress-mediated damage is often a downstream result of Parkinson's disease (PD), which is marked by sharp decline in dopaminergic neurons within the nigrostriatal regions of the brain, accounting for the symptomatic motor deficits in patients. Regulating the level of oxidative stress may present a beneficial approach in preventing PD pathology. Here, we assessed the efficacy of a nicotinamide adenine phosphate (NADPH) oxidase (NOX) inhibitor, an exogenous reactive oxygen species (ROS) regulator synthesized by Aptabio therapeutics with the specificity to NOX-1, 2 and 4. Utilizing N27 rat dopaminergic cells and C57Bl/6 mice, we confirmed that the exposures of alpha-synuclein preformed fibrils (PFF) induced protein aggregation, a hallmark in PD pathology. In vitro assessment of the novel compound revealed an increase in cell viability and decreases in cytotoxicity, ROS, and protein aggregation (Thioflavin-T stain) against PFF exposure at the optimal concentration of 10 nM. Concomitantly, the oral treatment alleviated motor-deficits in behavioral tests, such as hindlimb clasping, rotarod, pole, nesting and grooming test, via reducing protein aggregation, based on rescued dopaminergic neuronal loss. The suppression of NOX-1, 2 and 4 within the striatum and ventral midbrain regions including Substantia Nigra compacta (SNc) contributed to neuroprotective/recovery effects, making it a potential therapeutic option for PD.
Collapse
Affiliation(s)
- Kwadwo Ofori
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Anurupa Ghosh
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Dinesh Kumar Verma
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Darice Wheeler
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Gabriela Cabrera
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| | - Jong-Bok Seo
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Republic of Korea;
| | - Yong-Hwan Kim
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (K.O.); (A.G.); (D.K.V.); (D.W.); (G.C.)
| |
Collapse
|
10
|
Cipriano A, Viviano M, Feoli A, Milite C, Sarno G, Castellano S, Sbardella G. NADPH Oxidases: From Molecular Mechanisms to Current Inhibitors. J Med Chem 2023; 66:11632-11655. [PMID: 37650225 PMCID: PMC10510401 DOI: 10.1021/acs.jmedchem.3c00770] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Indexed: 09/01/2023]
Abstract
NADPH oxidases (NOXs) form a family of electron-transporting membrane enzymes whose main function is reactive oxygen species (ROS) generation. Strong evidence suggests that ROS produced by NOX enzymes are major contributors to oxidative damage under pathologic conditions. Therefore, blocking the undesirable actions of these enzymes is a therapeutic strategy for treating various pathological disorders, such as cardiovascular diseases, inflammation, and cancer. To date, identification of selective NOX inhibitors is quite challenging, precluding a pharmacologic demonstration of NOX as therapeutic targets in vivo. The aim of this Perspective is to furnish an updated outlook about the small-molecule NOX inhibitors described over the last two decades. Structures, activities, and in vitro/in vivo specificity are discussed, as well as the main biological assays used.
Collapse
Affiliation(s)
- Alessandra Cipriano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Monica Viviano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Alessandra Feoli
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Ciro Milite
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Giuliana Sarno
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Sabrina Castellano
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| | - Gianluca Sbardella
- Department
of Pharmacy, Epigenetic Med Chem Lab, and PhD Program in Drug Discovery and
Development, University of Salerno, via Giovanni Paolo II 132, I-84084 Fisciano, Salerno, Italy
| |
Collapse
|
11
|
Alves-Lopes R, Lacchini S, Neves KB, Harvey A, Montezano AC, Touyz RM. Vasoprotective effects of NOX4 are mediated via polymerase and transient receptor potential melastatin 2 cation channels in endothelial cells. J Hypertens 2023; 41:1389-1400. [PMID: 37272080 PMCID: PMC10399938 DOI: 10.1097/hjh.0000000000003478] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 05/16/2023] [Accepted: 05/17/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND NOX4 activation has been implicated to have vasoprotective and blood pressure (BP)-lowering effects. Molecular mechanisms underlying this are unclear, but NOX4-induced regulation of the redox-sensitive Ca 2+ channel TRPM2 and effects on endothelial nitric oxide synthase (eNOS)-nitric oxide signalling may be important. METHOD Wild-type and LinA3, renin-expressing hypertensive mice, were crossed with NOX4 knockout mice. Vascular function was measured by myography. Generation of superoxide (O 2- ) and hydrogen peroxide (H 2 O 2 ) were assessed by lucigenin and amplex red, respectively, and Ca 2+ influx by Cal-520 fluorescence in rat aortic endothelial cells (RAEC). RESULTS BP was increased in NOX4KO, LinA3 and LinA3/NOX4KO mice. This was associated with endothelial dysfunction and vascular remodelling, with exaggerated effects in NOX4KO groups. The TRPM2 activator, ADPR, improved vascular relaxation in LinA3/NOX4KO mice, an effect recapitulated by H 2 O 2 . Inhibition of PARP and TRPM2 with olaparib and 2-APB, respectively, recapitulated endothelial dysfunction in NOX4KO. In endothelial cells, Ang II increased H 2 O 2 generation and Ca 2+ influx, effects reduced by TRPM2 siRNA, TRPM2 inhibitors (8-br-cADPR, 2-APB), olaparib and GKT137831 (NOX4 inhibitor). Ang II-induced eNOS activation was blocked by NOX4 and TRPM2 siRNA, GKT137831, PEG-catalase and 8-br-cADPR. CONCLUSION Our findings indicate that NOX4-induced H 2 O 2 production activates PARP/TRPM2, Ca 2+ influx, eNOS activation and nitric oxide release in endothelial cells. NOX4 deficiency impairs Ca 2+ homeostasis leading to endothelial dysfunction, an effect exacerbated in hypertension. We define a novel pathway linking endothelial NOX4/H 2 O 2 to eNOS/nitric oxide through PARP/TRPM2/Ca 2+ . This vasoprotective pathway is perturbed when NOX4 is downregulated and may have significance in conditions associated with endothelial dysfunction, including hypertension.
Collapse
Affiliation(s)
- Rheure Alves-Lopes
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Silvia Lacchini
- Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Karla B. Neves
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, UK
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Adam Harvey
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Augusto C. Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Rhian M. Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Quebec, Canada
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| |
Collapse
|
12
|
Ali MY, Jannat S, Chang MS. Discovery of Potent Angiotensin-Converting Enzyme Inhibitors in Pomegranate as a Treatment for Hypertension. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023. [PMID: 37384918 DOI: 10.1021/acs.jafc.3c02115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/01/2023]
Abstract
Pomegranate (Punica granatum L.) is associated with numerous health benefits due to its high levels of antioxidant polyphenolic substances. Since pomegranate extract has been shown to inhibit angiotensin-converting enzyme (ACE), the potential inhibitory effect of most of its main constituents against ACE is unknown. Therefore, we tested the activities of 24 major compounds, the majority of which significantly inhibited ACE. Notably, pedunculagin, punicalin, and gallagic acid were the most effective ACE inhibitors with IC50 values of 0.91, 1.12, and 1.77 μM, respectively. As demonstrated in molecular docking studies, compounds block ACE by forming multiple hydrogen bonds and hydrophobic interactions with catalytic residues and zinc ions in ACE's C- and N-domains, consequently inhibiting ACE's catalytic activity. Also, the most active pedunculagin stimulated nitric oxide (NO) production, activated the endothelial nitric oxide synthase enzyme (eNOS), and significantly increased eNOS protein expression levels up to 5.3-fold in EA.hy926 cells. Furthermore, pedunculagin increased in cellular calcium (Ca2+) concentration promoted eNOS enzyme activation and reduced the production of reactive oxygen species (ROS). In addition, the active compounds improved glucose uptake in insulin-resistant C2C12 skeletal muscle cells in a dose-dependent manner. The results of these computational, in vitro, and cellular experiments provide further evidence to the traditional medicine that involves using pomegranates to treat cardiovascular diseases like hypertension.
Collapse
Affiliation(s)
- Md Yousof Ali
- Department of Clinical Neurosciences, University of Calgary, Calgary, AB T2N 1N4, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Susoma Jannat
- Department of Biochemistry and Molecular Biology, University of Calgary, Calgary, AB T2N 1N4, Canada
| | - Mun Seog Chang
- Department of Prescriptionology, College of Korean Medicine, Kyung Hee University, 26 Kyunghee dae-ro, Seoul 02447, Korea
- Qgenetics, Seoul Bio Cooperation Center 504, 23 Kyunghee dae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
13
|
Trostchansky A, Alarcon M. An Overview of Two Old Friends Associated with Platelet Redox Signaling, the Protein Disulfide Isomerase and NADPH Oxidase. Biomolecules 2023; 13:biom13050848. [PMID: 37238717 DOI: 10.3390/biom13050848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/30/2022] [Accepted: 01/18/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress participates at the baseline of different non-communicable pathologies such as cardiovascular diseases. Excessive formation of reactive oxygen species (ROS), above the signaling levels necessary for the correct function of organelles and cells, may contribute to the non-desired effects of oxidative stress. Platelets play a relevant role in arterial thrombosis, by aggregation triggered by different agonists, where excessive ROS formation induces mitochondrial dysfunction and stimulate platelet activation and aggregation. Platelet is both a source and a target of ROS, thus we aim to analyze both the platelet enzymes responsible for ROS generation and their involvement in intracellular signal transduction pathways. Among the proteins involved in these processes are Protein Disulphide Isomerase (PDI) and NADPH oxidase (NOX) isoforms. By using bioinformatic tools and information from available databases, a complete bioinformatic analysis of the role and interactions of PDI and NOX in platelets, as well as the signal transduction pathways involved in their effects was performed. We focused the study on analyzing whether these proteins collaborate to control platelet function. The data presented in the current manuscript support the role that PDI and NOX play on activation pathways necessary for platelet activation and aggregation, as well as on the platelet signaling imbalance produced by ROS production. Our data could be used to design specific enzyme inhibitors or a dual inhibition for these enzymes with an antiplatelet effect to design promising treatments for diseases involving platelet dysfunction.
Collapse
Affiliation(s)
- Andrés Trostchansky
- Departamento de Bioquímica and Center for Free Radical and Biomedical Research, Facultad de Medicina, Universidad de la República, Montevideo 11800, Uruguay
| | - Marcelo Alarcon
- Thrombosis Research Center, Universidad de Talca, Talca 3460000, Chile
- Department of Clinical Biochemistry and Immunohematology, Faculty of Health Sciences, Universidad de Talca, Talca 3460000, Chile
| |
Collapse
|
14
|
Briyal S, Ranjan AK, Gulati A. Oxidative stress: A target to treat Alzheimer's disease and stroke. Neurochem Int 2023; 165:105509. [PMID: 36907516 DOI: 10.1016/j.neuint.2023.105509] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 02/01/2023] [Accepted: 03/05/2023] [Indexed: 03/13/2023]
Abstract
Oxidative stress has been established as a well-known pathological condition in several neurovascular diseases. It starts with increased production of highly oxidizing free-radicals (e.g. reactive oxygen species; ROS and reactive nitrogen species; RNS) and becomes too high for the endogenous antioxidant system to neutralize them, which results in a significantly disturbed balance between free-radicals and antioxidants levels and causes cellular damage. A number of studies have evidently shown that oxidative stress plays a critical role in activating multiple cell signaling pathways implicated in both progression as well as initiation of neurological diseases. Therefore, oxidative stress continues to remain a key therapeutic target for neurological diseases. This review discusses the mechanisms involved in reactive oxygen species (ROS) generation in the brain, oxidative stress, and pathogenesis of neurological disorders such as stroke and Alzheimer's disease (AD) and the scope of antioxidant therapies for these disorders.
Collapse
Affiliation(s)
- Seema Briyal
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA.
| | - Amaresh K Ranjan
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA
| | - Anil Gulati
- College of Pharmacy, Midwestern University, Downers Grove, IL, 60515, USA; Pharmazz Inc. Research and Development, Willowbrook, IL, USA
| |
Collapse
|
15
|
Ruiz-Uribe NE, Bracko O, Swallow M, Omurzakov A, Dash S, Uchida H, Xiang D, Haft-Javaherian M, Falkenhain K, Lamont ME, Ali M, Njiru BN, Chang HY, Tan AY, Xiang JZ, Iadecola C, Park L, Sanchez T, Nishimura N, Schaffer CB. Vascular oxidative stress causes neutrophil arrest in brain capillaries, leading to decreased cerebral blood flow and contributing to memory impairment in a mouse model of Alzheimer’s disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.15.528710. [PMID: 36824768 PMCID: PMC9949082 DOI: 10.1101/2023.02.15.528710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
INTRODUCTION In this study, we explore the role of oxidative stress produced by NOX2-containing NADPH oxidase as a molecular mechanism causing capillary stalling and cerebral blood flow deficits in the APP/PS1 mouse model of AD. METHODS We inhibited NOX2 in APP/PS1 mice by administering a 10 mg/kg dose of the peptide inhibitor gp91-ds-tat i.p., for two weeks. We used in vivo two-photon imaging to measure capillary stalling, penetrating arteriole flow, and vascular inflammation. We also characterized short-term memory function and gene expression changes in cerebral microvessels. RESULTS We found that after NOX2 inhibition capillary stalling, as well as parenchymal and vascular inflammation, were significantly reduced. In addition, we found a significant increase in penetrating arteriole flow, followed by an improvement in short-term memory, and downregulation of inflammatory gene expression pathways. DISCUSSION Oxidative stress is a major mechanism leading to microvascular dysfunction in AD, and represents an important therapeutic target.
Collapse
|
16
|
Structure, Activation, and Regulation of NOX2: At the Crossroad between the Innate Immunity and Oxidative Stress-Mediated Pathologies. Antioxidants (Basel) 2023; 12:antiox12020429. [PMID: 36829988 PMCID: PMC9952346 DOI: 10.3390/antiox12020429] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) is a multisubunit enzyme complex that participates in the generation of superoxide or hydrogen peroxide (H2O2) and plays a key role in several biological functions. Among seven known NOX isoforms, NOX2 was the first identified in phagocytes but is also expressed in several other cell types including endothelial cells, platelets, microglia, neurons, and muscle cells. NOX2 has been assigned multiple roles in regulating many aspects of innate and adaptive immunity, and human and mouse models of NOX2 genetic deletion highlighted this key role. On the other side, NOX2 hyperactivation is involved in the pathogenesis of several diseases with different etiologies but all are characterized by an increase in oxidative stress and inflammatory process. From this point of view, the modulation of NOX2 represents an important therapeutic strategy aimed at reducing the damage associated with its hyperactivation. Although pharmacological strategies to selectively modulate NOX2 are implemented thanks to new biotechnologies, this field of research remains to be explored. Therefore, in this review, we analyzed the role of NOX2 at the crossroads between immunity and pathologies mediated by its hyperactivation. We described (1) the mechanisms of activation and regulation, (2) human, mouse, and cellular models studied to understand the role of NOX2 as an enzyme of innate immunity, (3) some of the pathologies associated with its hyperactivation, and (4) the inhibitory strategies, with reference to the most recent discoveries.
Collapse
|
17
|
Chu YC, Fang HW, Wu YY, Tang YJ, Hsieh EH, She Y, Chang CY, Lin IC, Chen YJ, Liu GS, Tseng CL. Functional Peptide-Loaded Gelatin Nanoparticles as Eyedrops for Cornea Neovascularization Treatment. Int J Nanomedicine 2023; 18:1413-1431. [PMID: 36992821 PMCID: PMC10042260 DOI: 10.2147/ijn.s398769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/07/2023] [Indexed: 03/31/2023] Open
Abstract
Background Corneal neovascularization (NV) is a process of abnormal vessel growth into the transparent cornea from the limbus and can disturb the light passing through the cornea, resulting in vision loss or even blindness. The use of nanomedicine as an effective therapeutic formulation in ophthalmology has led to higher drug bioavailability and a slow drug release rate. In this research, we designed and explored the feasibility of a new nanomedicine, gp91 ds-tat (gp91) peptide-encapsulated gelatin nanoparticles (GNP-gp91), for inhibiting corneal angiogenesis. Methods GNP-gp91 were prepared by a two-step desolvation method. The characterization and cytocompatibility of GNP-gp91 were analyzed. The inhibition effect of GNP-gp91 on HUVEC cell migration and tube formation was observed by an inverted microscope. The drug retention test in mouse cornea was observed by in vivo imaging system, fluorescence microscope, and DAPI/TAMRA staining. Finally, the therapeutic efficacy and evaluation of neovascularization-related factors were conducted through the in vivo corneal NV mice model via topical delivery. Results The prepared GNP-gp91 had a nano-scale diameter (550.6 nm) with positive charge (21.7 mV) slow-release behavior (25%, 240hr). In vitro test revealed that GNP-gp91 enhanced the inhibition of cell migration and tube formation capacity via higher internalization of HUVEC. Topical administration (eyedrops) of the GNP-gp91 significantly prolongs the retention time (46%, 20 min) in the mouse cornea. In chemically burned corneal neovascularization models, corneal vessel area with a significant reduction in GNP-gp91 group (7.89%) was revealed when compared with PBS (33.99%) and gp91 (19.67%) treated groups via every two days dosing. Moreover, GNP-gp91 significantly reduced the concentration of Nox2, VEGF and MMP9 in NV's cornea. Conclusion The nanomedicine, GNP-gp91, was successfully synthesized for ophthalmological application. These data suggest that GNP-gp91 contained eyedrops that not only have a longer retention time on the cornea but also can treat mice corneal NV effectively delivered in a low dosing frequency, GNP-gp91 eyedrops provides an alternative strategy for clinical ocular disease treatment in the culture.
Collapse
Affiliation(s)
- Ya-Chun Chu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei City, Taiwan
| | - Hsu-Wei Fang
- Department of Chemical Engineering and Biotechnology, National Taipei University of Technology, Taipei City, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli County, Taiwan
| | - Yu-Yi Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
| | - Yu-Jun Tang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
| | - Erh-Hsuan Hsieh
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
| | - YiZhou She
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
| | - Che-Yi Chang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
| | - I-Chan Lin
- Department of Ophthalmology, Wan Fang Hospital, Taipei Medical University, Taipei City, Taiwan
- Department of Ophthalmology, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei City, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
- Center for Precision Medicine and Translational Cancer Research, Taipei Medical University Hospital, Taipei City, Taiwan
| | - Guei-Sheung Liu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, Australia
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Ching-Li Tseng
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
- Center for Precision Medicine and Translational Cancer Research, Taipei Medical University Hospital, Taipei City, Taiwan
- Research Center of Biomedical Device, College of Biomedical Engineering, Taipei Medical University, Taipei City, Taiwan
- Correspondence: Ching-Li Tseng, Tel +886 2 2736 1661 (ext. 5214), Email
| |
Collapse
|
18
|
Singh PK, Saadi A, Sheeni Y, Shekh-Ahmad T. Specific inhibition of NADPH oxidase 2 modifies chronic epilepsy. Redox Biol 2022; 58:102549. [PMID: 36459714 PMCID: PMC9712695 DOI: 10.1016/j.redox.2022.102549] [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: 09/01/2022] [Revised: 10/29/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
Recent work by us and others has implicated NADPH oxidase (NOX) enzymes as main producers of reactive oxygen species (ROS) following a brain insult such as status epilepticus, contributing to neuronal damage and development of epilepsy. Although several NOX isoforms have been examined in the context of epilepsy, most attention has focused on NOX2. In this present study, we demonstrate the effect of gp91ds-tat, a specific competitive inhibitor of NOX2, in in vitro epileptiform activity model as well as in temporal lobe epilepsy (TLE) model in rats. We showed that in in vitro seizure model, gp91ds-tat modulated Ca2+ oscillation, prevented epileptiform activity-induced ROS generation, mitochondrial depolarization, and neuronal death. Administration of gp91ds-tat 1 h after kainic acid-induced status epilepticus significantly decreased the expression of NOX2, as well as the overall NOX activity in the cortex and the hippocampus. Finally, we showed that upon continuous intracerebroventricular administration to epileptic rats, gp91ds-tat significantly reduced the seizure frequency and the total number of seizures post-treatment compared to the scrambled peptide-treated animals. The results of the study suggest that NOX2 may have an important effect on modulation of epileptiform activity and has a critical role in mediating seizure-induced NOX activation, ROS generation and oxidative stress in the brain, and thus significantly contributes to development of epilepsy following a brain insult.
Collapse
Affiliation(s)
| | | | | | - Tawfeeq Shekh-Ahmad
- Corresponding author. The Institute for Drug Research, The School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
| |
Collapse
|
19
|
Keeney MT, Hoffman EK, Farmer K, Bodle CR, Fazzari M, Zharikov A, Castro SL, Hu X, Mortimer A, Kofler JK, Cifuentes-Pagano E, Pagano PJ, Burton EA, Hastings TG, Greenamyre JT, Di Maio R. NADPH oxidase 2 activity in Parkinson's disease. Neurobiol Dis 2022; 170:105754. [PMID: 35577065 PMCID: PMC9284948 DOI: 10.1016/j.nbd.2022.105754] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022] Open
Abstract
Mitochondrial dysfunction and oxidative stress are strongly implicated in Parkinson's disease (PD) pathogenesis and there is evidence that mitochondrially-generated superoxide can activate NADPH oxidase 2 (NOX2). Although NOX2 has been examined in the context of PD, most attention has focused on glial NOX2, and the role of neuronal NOX2 in PD remains to be defined. Additionally, pharmacological NOX2 inhibitors have typically lacked specificity. Here we devised and validated a proximity ligation assay for NOX2 activity and demonstrated that in human PD and two animal models thereof, both neuronal and microglial NOX2 are highly active in substantia nigra under chronic conditions. However, in acute and sub-acute PD models, we observed neuronal, but not microglial NOX2 activation, suggesting that neuronal NOX2 may play a primary role in the early stages of the disease. Aberrant NOX2 activity is responsible for the formation of oxidative stress-related post-translational modifications of α-synuclein, and impaired mitochondrial protein import in vitro in primary ventral midbrain neuronal cultures and in vivo in nigrostriatal neurons in rats. In a rat model, administration of a brain-penetrant, highly specific NOX2 inhibitor prevented NOX2 activation in nigrostriatal neurons and its downstream effects in vivo, such as activation of leucine-rich repeat kinase 2 (LRRK2). We conclude that NOX2 is an important enzyme that contributes to progressive oxidative damage which in turn can lead to α-synuclein accumulation, mitochondrial protein import impairment, and LRRK2 activation. In this context, NOX2 inhibitors hold potential as a disease-modifying therapy in PD.
Collapse
Affiliation(s)
- Matthew T Keeney
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eric K Hoffman
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kyle Farmer
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Christopher R Bodle
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alevtina Zharikov
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sandra L Castro
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaoping Hu
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Amanda Mortimer
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Julia K Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Patrick J Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Teresa G Hastings
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Roberto Di Maio
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
20
|
Stanhewicz AE, Schlarmann RL, Brustkern KM, Jalal D. Oxidative stress contributes to reductions in microvascular endothelial- and nitric oxide-dependent dilation in women with a history of gestational diabetes. J Appl Physiol (1985) 2022; 133:361-370. [PMID: 35796611 PMCID: PMC9359638 DOI: 10.1152/japplphysiol.00189.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Women with a history of gestational diabetes mellitus (GDM) are twice as likely to develop cardiovascular disease and ~7x as likely to develop type II diabetes as their age-matched counterparts. However, the mechanism(s) mediating these associations remain unclear. We hypothesized that endothelium- and NO-dependent dilation would be attenuated through oxidant stress mechanisms in the microvasculature of women with a history of GDM compared to control women with a history of uncomplicated pregnancy (HC). Ten HC (35±4yrs) and 10 GDM (34±4yrs) underwent a standard local heating protocol (42°C; 0.1°C·s-1). Two intradermal microdialysis fibers were placed in the ventral forearm for local delivery of lactated Ringer's (control), or 5mM L-ascorbate. After full expression of the local heating response, 15mM NG-nitro-L-arginine methyl ester NO synthase-inhibition) was perfused. Red cell flux was measured continuously by laser-Doppler flowmetry and cutaneous vascular conductance (CVC=flux/MAP) was standardized to maximum (%CVCmax; 28mM SNP + 43°C). Urine albumin:creatinine ratio (ACR) was measured. GDM had attenuated endothelium-dependent (GDM: 67±7 vs. HC: 90±4%CVCmax; p<0.001) and NO-dependent (GDM: 54±7 vs. HC: 71±3%; p=0.001) dilation at the control site and tended to have higher urine ACR (p=0.06). Both endothelium-dependent (r2=0.53, p=0.02) and NO-dependent (r2=0.56, p=0.01) dilation were related to urine ACR in GDM. L-ascorbate perfusion improved endothelium-dependent (82±5%CVCmax; p=0.03 vs. control) and NO-dependent (68±5%; p=0.02 vs. control) dilation in GDM but had no effect in HC (p>0.05). Otherwise healthy women with a history of GDM have attenuated microvascular endothelial function and this dysfunction is mediated, in part, by oxidative stress.
Collapse
Affiliation(s)
- Anna E Stanhewicz
- Department of Health and Human Physiology, The University of Iowa, Iowa City, Iowa, United States
| | - Rowan L Schlarmann
- Department of Health and Human Physiology, The University of Iowa, Iowa City, Iowa, United States
| | - Kaila M Brustkern
- Department of Health and Human Physiology, The University of Iowa, Iowa City, Iowa, United States
| | - Diana Jalal
- The Iowa City VA HCS, Iowa City, IA, United States.,Department of Internal Medicine, Carver College of Medicine, Iowa City, IA, United States
| |
Collapse
|
21
|
Ghosh AA, Verma DK, Cabrera G, Ofori K, Hernandez-Quijada K, Kim JK, Chung JH, Moore M, Moon SH, Seo JB, Kim YH. A Novel NOX Inhibitor Treatment Attenuates Parkinson's Disease-Related Pathology in Mouse Models. Int J Mol Sci 2022; 23:4262. [PMID: 35457082 PMCID: PMC9030373 DOI: 10.3390/ijms23084262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/04/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative motor disorder without an available therapeutic to halt the formation of Lewy bodies for preventing dopaminergic neuronal loss in the nigrostriatal pathway. Since oxidative-stress-mediated damage has been commonly reported as one of the main pathological mechanisms in PD, we assessed the efficacy of a novel NOX inhibitor from AptaBio Therapeutics (C-6) in dopaminergic cells and PD mouse models. The compound reduced the cytotoxicity and enhanced the cell viability at various concentrations against MPP+ and α-synuclein preformed fibrils (PFFs). Further, the levels of ROS and protein aggregation were significantly reduced at the optimal concentration (1 µM). Using two different mouse models, we gavaged C-6 at two different doses to the PD sign-displaying transgenic mice for 2 weeks and stereotaxically PFF-injected mice for 5 weeks. Our results demonstrated that both C-6-treated mouse models showed alleviated motor deficits in pole test, hindlimb clasping, crossbeam, rotarod, grooming, and nesting analyses. We also confirmed that the compound treatment reduced the levels of protein aggregation, along with phosphorylated-α-synuclein, in the striatum and ventral midbrain and further dopaminergic neuronal loss. Taken together, our results strongly suggest that NOX inhibition can be a potential therapeutic target for PD.
Collapse
Affiliation(s)
- Anurupa A. Ghosh
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Dinesh Kumar Verma
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Gabriela Cabrera
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Kwadwo Ofori
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Karina Hernandez-Quijada
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| | - Jae-Kwan Kim
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Korea; (J.-K.K.); (J.H.C.); (J.B.S.)
| | - Joo Hee Chung
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Korea; (J.-K.K.); (J.H.C.); (J.B.S.)
| | - Michael Moore
- Imaging Core, Delaware State University, Dover, DE 19901, USA;
| | - Sung Hwan Moon
- AptaBio Therapeutics Inc., 504 Tower, Heungdeok IT Valley, Heungdeok 1-ro 13, Gyeonggi-do, Yongin-si 16954, Korea;
| | - Jong Bok Seo
- Seoul Center, Korea Basic Science Institute, Seongbuk-gu, Seoul 02841, Korea; (J.-K.K.); (J.H.C.); (J.B.S.)
| | - Yong-Hwan Kim
- Department of Biological Sciences/Neuroscience Program, Delaware State University, Dover, DE 19901, USA; (D.K.V.); (G.C.); (K.O.); (K.H.-Q.)
| |
Collapse
|
22
|
Deficiency of NARFL increases transcription of NADPH oxidases and ROS production impairing the function of endothelial cells. Life Sci 2022; 301:120567. [DOI: 10.1016/j.lfs.2022.120567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/27/2022] [Accepted: 04/18/2022] [Indexed: 11/20/2022]
|
23
|
Jurcau A, Ardelean AI. Oxidative Stress in Ischemia/Reperfusion Injuries following Acute Ischemic Stroke. Biomedicines 2022; 10:biomedicines10030574. [PMID: 35327376 PMCID: PMC8945353 DOI: 10.3390/biomedicines10030574] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/25/2022] [Accepted: 02/28/2022] [Indexed: 02/04/2023] Open
Abstract
Recanalization therapy is increasingly used in the treatment of acute ischemic stroke. However, in about one third of these patients, recanalization is followed by ischemia/reperfusion injuries, and clinically to worsening of the neurological status. Much research has focused on unraveling the involved mechanisms in order to prevent or efficiently treat these injuries. What we know so far is that oxidative stress and mitochondrial dysfunction are significantly involved in the pathogenesis of ischemia/reperfusion injury. However, despite promising results obtained in experimental research, clinical studies trying to interfere with the oxidative pathways have mostly failed. The current article discusses the main mechanisms leading to ischemia/reperfusion injuries, such as mitochondrial dysfunction, excitotoxicity, and oxidative stress, and reviews the clinical trials with antioxidant molecules highlighting recent developments and future strategies.
Collapse
Affiliation(s)
- Anamaria Jurcau
- Department of Psycho-Neurosciences and Rehabilitation, Faculty of Medicine and Pharmacy, University of Oradea, 410087 Oradea, Romania
- Department of Neurology, Clinical Municipal Hospital Oradea, Louis Pasteur Street nr 26, 410054 Oradea, Romania
- Correspondence: ; Tel.: +40-744-600-833
| | - Adriana Ioana Ardelean
- Department of Preclinical Sciences, Faculty of Medicine and Pharmacy, University of Oradea, Universitatii Street nr 1, 410087 Oradea, Romania;
- Department of Cardiology, Clinical Emergency County Hospital Oradea, Gh. Doja Street nr 65, 410169 Oradea, Romania
| |
Collapse
|
24
|
Alwjwaj M, Kadir RRA, Bayraktutan U. Outgrowth endothelial progenitor cells restore cerebral barrier function following ischaemic damage: the impact of NOX2 inhibition. Eur J Neurosci 2022; 55:1658-1670. [PMID: 35179812 DOI: 10.1111/ejn.15627] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 02/08/2022] [Accepted: 02/15/2022] [Indexed: 11/29/2022]
Abstract
Disruption of blood-brain barrier (BBB), formed mainly by human brain microvascular endothelial cells (HBMECs), constitutes the major cause of mortality following ischaemic stroke. This study investigates whether OECs (outgrowth endothelial cells) can restore BBB integrity and function following ischaemic damage, and how inhibition of NOX2, a main source of vascular oxidative stress, affects the characteristics of BBB established with OECs and HBMECs in ischaemic settings. In vitro models of human BBB were constructed by co-culture of pericytes and astrocytes with either OECs or HBMECs before exposure to oxygen-glucose deprivation (OGD) alone or followed by reperfusion (OGD+R) in the absence or presence of NOX2 inhibitor, gp91ds-tat. The function and integrity of BBB were assessed by measurements of paracellular flux of sodium fluorescein (NaF) and transendothelial electrical resistance (TEER), respectively. Treatment with OECs during OGD+R effectively restored BBB integrity and function. Compared to HBMECs, OECs possessed lower NADPH oxidase activity, superoxide anion levels, and had greater total antioxidant capacity during OGD and OGD+R. Inhibition of NADPH oxidase during OGD and OGD+R restored the integrity and function of BBB established by HBMECs. This was evidenced by reductions in NADPH oxidase activity and superoxide anion levels. In contrast, treatment with gp91ds-tat aggravated ischaemic injury-induced BBB damage constructed by OECs. In conclusion, OECs are more resistant to ischaemic conditions and can effectively repair cerebral barrier following ischaemic damage. Suppression of oxidative stress through specific targeting of NOX2 requires close attention while using OECs as therapeutics.
Collapse
Affiliation(s)
- Mansour Alwjwaj
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, Nottingham, UK
| | - Rais Reskiawan A Kadir
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, Nottingham, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, Nottingham, UK
| |
Collapse
|
25
|
Jiang L, Li Y, Wang L, Guo J, Liu W, Meng G, Zhang L, Li M, Cong L, Sun M. Recent Insights Into the Prognostic and Therapeutic Applications of Lysozymes. Front Pharmacol 2021; 12:767642. [PMID: 34925025 PMCID: PMC8678502 DOI: 10.3389/fphar.2021.767642] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 11/10/2021] [Indexed: 01/15/2023] Open
Abstract
Lysozymes are naturally occurring enzymes present in a variety of biological organisms, such as bacteria, fungi, and animal bodily secretions and tissues. It is also the main ingredient of many ethnomedicines. It is well known that lysozymes and lysozyme-like enzymes can be used as anti-bacterial agents by degrading bacterial cell wall peptidoglycan that leads to cell death, and can also inhibit fungi, yeasts, and viruses. In addition to its direct antimicrobial activity, lysozyme is also an important component of the innate immune system in most mammals. Increasing evidence has shown the immune-modulatory effects of lysozymes against infection and inflammation. More recently, studies have revealed the anti-cancer activities of lysozyme in multiple types of tumors, potentially through its immune-modulatory activities. In this review, we summarized the major functions and underlying mechanisms of lysozymes derived from animal and plant sources. We highlighted the therapeutic applications and recent advances of lysozymes in cancers, hypertension, and viral diseases, aiming toseeking alternative therapies for standard medical treatment bypassing side effects. We also evaluated the role of lysozyme as a promising cancer marker for prognosis to indicate the outcomes recurrence for patients.
Collapse
Affiliation(s)
- Lin Jiang
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Yunhe Li
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Liye Wang
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, United States
| | - Jian Guo
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Wei Liu
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Guixian Meng
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Lei Zhang
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| | - Miao Li
- Department of Neurosurgery, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lina Cong
- School of Biological Engineering, Dalian Polytechnic University, Dalian, China
| | - Meiyan Sun
- College of Laboratory Medicine, Jilin Medical University, Jilin, China
| |
Collapse
|
26
|
Villegas L, Nørremølle A, Freude K, Vilhardt F. Nicotinamide Adenine Dinucleotide Phosphate Oxidases Are Everywhere in Brain Disease, but Not in Huntington's Disease? Front Aging Neurosci 2021; 13:736734. [PMID: 34803655 PMCID: PMC8602359 DOI: 10.3389/fnagi.2021.736734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder characterized by neuronal loss and tissue atrophy mainly in the striatum and cortex. In the early stages of the disease, impairment of neuronal function, synaptic dysfunction and white matter loss precedes neuronal death itself. Relative to other neurodegenerative diseases such as Alzheimer's and Parkinson's disease and Amyotrophic Lateral Sclerosis, where the effects of either microglia or NADPH oxidases (NOXs) are recognized as important contributors to disease pathogenesis and progression, there is a pronounced lack of information in HD. This information void contrasts with evidence from human HD patients where blood monocytes and microglia are activated well before HD clinical symptoms (PET scans), and the clear signs of oxidative stress and inflammation in post mortem HD brain. Habitually, NOX activity and oxidative stress in the central nervous system (CNS) are equated with microglia, but research of the last two decades has carved out important roles for NOX enzyme function in neurons. Here, we will convey recent information about the function of NOX enzymes in neurons, and contemplate on putative roles of neuronal NOX in HD. We will focus on NOX-produced reactive oxygen species (ROS) as redox signaling molecules in/among neurons, and the specific roles of NOXs in important processes such as neurogenesis and lineage specification, neurite outgrowth and growth cone dynamics, and synaptic plasticity where NMDAR-dependent signaling, and long-term depression/potentiation are redox-regulated phenomena. HD animal models and induced pluripotent stem cell (iPSC) studies have made it clear that the very same physiological processes are also affected in HD, and we will speculate on possible roles for NOX in the pathogenesis and development of disease. Finally, we also take into account the limited information on microglia in HD and relate this to any contribution of NOX enzymes.
Collapse
Affiliation(s)
- Luisana Villegas
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Anne Nørremølle
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Kristine Freude
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Fang J, Sheng R, Qin ZH. NADPH Oxidases in the Central Nervous System: Regional and Cellular Localization and the Possible Link to Brain Diseases. Antioxid Redox Signal 2021; 35:951-973. [PMID: 34293949 DOI: 10.1089/ars.2021.0040] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Significance: The significant role of reduced nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (Nox) in signal transduction is mediated by the production of reactive oxygen species (ROS), especially in the central nervous system (CNS). The pathogenesis of some neurologic and psychiatric diseases is regulated by ROS, acting as a second messenger or pathogen. Recent Advances: In the CNS, the involvement of Nox-derived ROS has been implicated in the regulation of multiple signals, including cell survival/apoptosis, neuroinflammation, migration, differentiation, proliferation, and synaptic plasticity, as well as the integrity of the blood/brain barrier. In these processes, the intracellular signals mediated by the members of the Nox family vary among different tissues. The present review illuminates the regions and cellular, subcellular localization of Nox isoforms in the brain, the signal transduction, and the role of NOX enzymes in pathophysiology, respectively. Critical Issues: Different signal transduction cascades are coupled to ROS derived from various Nox homologues with varying degrees. Therefore, a critical issue worth noting is the varied role of the homologues of NOX enzymes in different signaling pathways and also they mediate different phenotypes in the diverse pathophysiological condition. This substantiates the effectiveness of selective Nox inhibitors in the CNS. Future Directions: Further investigation to elucidate the role of various homologues of NOX enzymes in acute and chronic brain diseases and signaling mechanisms, and the development of more specific NOX inhibitors for the treatment of CNS disease are urgently needed. Antioxid. Redox Signal. 35, 951-973.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, China
| |
Collapse
|
28
|
Taylor JP, Tse HM. The role of NADPH oxidases in infectious and inflammatory diseases. Redox Biol 2021; 48:102159. [PMID: 34627721 PMCID: PMC8487856 DOI: 10.1016/j.redox.2021.102159] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/30/2021] [Accepted: 09/30/2021] [Indexed: 02/06/2023] Open
Abstract
Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOX) are enzymes that generate superoxide or hydrogen peroxide from molecular oxygen utilizing NADPH as an electron donor. There are seven enzymes in the NOX family: NOX1-5 and dual oxidase (DUOX) 1-2. NOX enzymes in humans play important roles in diverse biological functions and vary in expression from tissue to tissue. Importantly, NOX2 is involved in regulating many aspects of innate and adaptive immunity, including regulation of type I interferons, the inflammasome, phagocytosis, antigen processing and presentation, and cell signaling. DUOX1 and DUOX2 play important roles in innate immune defenses at epithelial barriers. This review discusses the role of NOX enzymes in normal physiological processes as well as in disease. NOX enzymes are important in autoimmune diseases like type 1 diabetes and have also been implicated in acute lung injury caused by infection with SARS-CoV-2. Targeting NOX enzymes directly or through scavenging free radicals may be useful therapies for autoimmunity and acute lung injury where oxidative stress contributes to pathology.
Collapse
Affiliation(s)
- Jared P Taylor
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hubert M Tse
- Department of Microbiology, Comprehensive Diabetes Center, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
29
|
Kadir RRA, Alwjwaj M, McCarthy Z, Bayraktutan U. Therapeutic hypothermia augments the restorative effects of PKC-β and Nox2 inhibition on an in vitro model of human blood-brain barrier. Metab Brain Dis 2021; 36:1817-1832. [PMID: 34398388 PMCID: PMC8437893 DOI: 10.1007/s11011-021-00810-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 07/26/2021] [Indexed: 12/24/2022]
Abstract
To investigate whether therapeutic hypothermia augments the restorative impact of protein kinase C-β (PKC-β) and Nox2 inhibition on an in vitro model of human blood-brain barrier (BBB). Cells cultured in normoglycaemic (5.5 mM) or hyperglycaemic (25 mM, 6 to 120 h) conditions were treated with therapeutic hypothermia (35 °C) in the absence or presence of a PKC-β inhibitor (LY333531, 0.05 μM) or a Nox2 inhibitor (gp91ds-tat, 50 μM). BBB was established by co-culture of human brain microvascular endothelial cells (HBMECs) with astrocytes (HAs) and pericytes. BBB integrity and function were assessed via transendothelial electrical resistance (TEER) and paracellular flux of sodium fluorescein (NaF, 376 Da). Nox activity (lucigenin assay), superoxide anion production (cytochrome-C reduction assay), cellular proliferative capacity (wound scratch assay) and actin cytoskeletal formation (rhodamine-phalloidin staining) were assessed both in HBMECs and HAs using the specific methodologies indicated in brackets. Therapeutic hypothermia augmented the protective effects of PKC-β or Nox2 inhibition on BBB integrity and function in experimental setting of hyperglycaemia, as evidenced by increases in TEER and concomitant decreases in paracellular flux of NaF. The combinatory approaches were more effective in repairing physical damage exerted on HBMEC and HA monolayers by wound scratch and in decreasing Nox activity and superoxide anion production compared to sole treatment regimen with either agent. Similarly, the combinatory approaches were more effective in suppressing actin stress fibre formation and maintaining normal cytoskeletal structure. Therapeutic hypothermia augments the cerebral barrier-restorative capacity of agents specifically targeting PKC-β or Nox2 pathways.
Collapse
Affiliation(s)
- Rais Reskiawan A Kadir
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Mansour Alwjwaj
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Zoe McCarthy
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK
| | - Ulvi Bayraktutan
- Academic Unit of Mental Health and Clinical Neuroscience, School of Medicine, The University of Nottingham, Hucknall Road, Nottingham, NG5 1PB, UK.
| |
Collapse
|
30
|
Trevelin SC, Sag CM, Zhang M, Alves-Filho JC, Cunha TM, dos Santos CX, Sawyer G, Murray T, Brewer A, Laurindo FRM, Protti A, Lopes LR, Ivetic A, Cunha FQ, Shah AM. Endothelial Nox2 Limits Systemic Inflammation and Hypotension in Endotoxemia by Controlling Expression of Toll-Like Receptor 4. Shock 2021; 56:268-277. [PMID: 34276040 PMCID: PMC8284354 DOI: 10.1097/shk.0000000000001706] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/29/2020] [Accepted: 12/02/2020] [Indexed: 02/01/2023]
Abstract
ABSTRACT Leukocyte Nox2 is recognized to have a fundamental microbicidal function in sepsis but the specific role of Nox2 in endothelial cells (EC) remains poorly elucidated. Here, we tested the hypothesis that endothelial Nox2 participates in the pathogenesis of systemic inflammation and hypotension induced by LPS. LPS was injected intravenously in mice with Tie2-targeted deficiency or transgenic overexpression of Nox2. Mice with Tie2-targeted Nox2 deficiency had increased circulating levels of TNF-α, enhanced numbers of neutrophils trapped in lungs, and aggravated hypotension after LPS injection, as compared to control LPS-injected animals. In contrast, Tie2-driven Nox2 overexpression attenuated inflammation and prevented the hypotension induced by LPS. Because Tie2-Cre targets both EC and myeloid cells we generated bone marrow chimeric mice with Nox2 deletion restricted to leukocytes or ECs. Mice deficient in Nox2 either in leukocytes or ECs had reduced LPS-induced neutrophil trapping in the lungs and lower plasma TNF-α levels as compared to control LPS-injected mice. However, the pronounced hypotensive response to LPS was present only in mice with EC-specific Nox2 deletion. Experiments in vitro with human vein or aortic endothelial cells (HUVEC and HAEC, respectively) treated with LPS revealed that EC Nox2 controls NF-κB activation and the transcription of toll-like receptor 4 (TLR4), which is the recognition receptor for LPS. In conclusion, these results suggest that endothelial Nox2 limits NF-κB activation and TLR4 expression, which in turn attenuates the severity of hypotension and systemic inflammation induced by LPS.
Collapse
Affiliation(s)
- Silvia Cellone Trevelin
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Can Martin Sag
- Department of Internal Medicine II, University Hospital of Regensburg, Regensburg, Germany
| | - Min Zhang
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - José Carlos Alves-Filho
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Thiago Mattar Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Célio Xavier dos Santos
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Greta Sawyer
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Thomas Murray
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Alison Brewer
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | | | - Andrea Protti
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Lucia Rossetti Lopes
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Aleksandar Ivetic
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| | - Fernando Queiroz Cunha
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, Brazil
| | - Ajay M. Shah
- King's College London, British Heart Foundation Centre for Research Excellence, School of Cardiovascular Medicine & Sciences, London, UK
| |
Collapse
|
31
|
Inoue M, Sakamoto K, Suzuki A, Nakai S, Ando A, Shiraki Y, Nakahara Y, Omura M, Enomoto A, Nakase I, Sawada M, Hashimoto N. Size and surface modification of silica nanoparticles affect the severity of lung toxicity by modulating endosomal ROS generation in macrophages. Part Fibre Toxicol 2021; 18:21. [PMID: 34134732 PMCID: PMC8210371 DOI: 10.1186/s12989-021-00415-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 05/25/2021] [Indexed: 11/10/2022] Open
Abstract
Background As the application of silica nanomaterials continues to expand, increasing chances of its exposure to the human body and potential harm are anticipated. Although the toxicity of silica nanomaterials is assumed to be affected by their physio-chemical properties, including size and surface functionalization, its molecular mechanisms remain unclear. We hypothesized that analysis of intracellular localization of the particles and subsequent intracellular signaling could reveal a novel determinant of inflammatory response against silica particles with different physico-chemical properties. Results We employed a murine intratracheal instillation model of amorphous silica nanoparticles (NPs) exposure to compare their in vivo toxicities in the respiratory system. Pristine silica-NPs of 50 nm diameters (50 nm-plain) induced airway-centered lung injury with marked neutrophilic infiltration. By contrast, instillation of pristine silica particles of a larger diameter (3 μm; 3 μm-plain) significantly reduced the severity of lung injury and neutrophilic infiltration, possibly through attenuated induction of neutrophil chemotactic chemokines including MIP2. Ex vivo analysis of alveolar macrophages as well as in vitro assessment using RAW264.7 cells revealed a remarkably lower cellular uptake of 3 μm-plain particles compared with 50 nm-plain, which is assumed to be the underlying mechanism of attenuated immune response. The severity of lung injury and neutrophilic infiltration was also significantly reduced after intratracheal instillation of silica NPs with an amine surface modification (50 nm-NH2) when compared with 50 nm-plain. Despite unchanged efficacy in cellular uptake, treatment with 50 nm-NH2 induced a significantly attenuated immune response in RAW264.7 cells. Assessment of intracellular redox signaling revealed increased reactive oxygen species (ROS) in endosomal compartments of RAW264.7 cells treated with 50 nm-plain when compared with vehicle-treated control. In contrast, augmentation of endosomal ROS signals in cells treated with 50 nm-NH2 was significantly lower. Moreover, selective inhibition of NADPH oxidase 2 (NOX2) was sufficient to inhibit endosomal ROS bursts and induction of chemokine expressions in cells treated with silica NPs, suggesting the central role of endosomal ROS generated by NOX2 in the regulation of the inflammatory response in macrophages that endocytosed silica NPs. Conclusions Our murine model suggested that the pulmonary toxicity of silica NPs depended on their physico-chemical properties through distinct mechanisms. Cellular uptake of larger particles by macrophages decreased, while surface amine modification modulated endosomal ROS signaling via NOX2, both of which are assumed to be involved in mitigating immune response in macrophages and resulting lung injury. Supplementary Information The online version contains supplementary material available at 10.1186/s12989-021-00415-0.
Collapse
Affiliation(s)
- Masahide Inoue
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Koji Sakamoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Atsushi Suzuki
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shinya Nakai
- Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, 599-8570, Japan
| | - Akira Ando
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yukihiko Shiraki
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoshio Nakahara
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Mika Omura
- Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, 599-8570, Japan
| | - Atsushi Enomoto
- Department of Pathology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ikuhiko Nakase
- Graduate School of Science, Osaka Prefecture University, Sakai, Osaka, 599-8570, Japan
| | - Makoto Sawada
- Department of Brain Function, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan.,Department of Molecular Pharmacokinetics Graduate School of Medicine, Nagoya University, Nagoya, Japan
| | - Naozumi Hashimoto
- Department of Respiratory Medicine, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| |
Collapse
|
32
|
Ling QS, Zhang SL, Tian JS, Cheng MH, Liu AJ, Fu FH, Liu JG, Miao CY. Allisartan isoproxil reduces mortality of stroke-prone rats and protects against cerebrovascular, cardiac, and aortic damage. Acta Pharmacol Sin 2021; 42:871-884. [PMID: 34002042 PMCID: PMC8149727 DOI: 10.1038/s41401-021-00684-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 04/17/2021] [Indexed: 12/22/2022] Open
Abstract
Stroke is a common cause of death and disability. Allisartan isoproxil (ALL) is a new angiotensin II receptor blocker and a new antihypertensive drug discovered and developed in China. In the present study we investigated the therapeutic effects of ALL in stroke-prone renovascular hypertensive rats (RHR-SP) and the underlying mechanisms. The model rats were generated via two-kidney two-clip (2K2C) surgery, which led to 100% of hypertension, 100% of cerebrovascular damage as well as 100% of mortality 1 year after the surgery. Administration of ALL (30 mg · kg-1 · d-1 in diet, for 55 weeks) significantly decreased stroke-related death and prolonged lifespan in RHR-SP, but the survival ALL-treated RHR-SP remained of hypertension and cardiovascular hypertrophy compared with sham-operated normal controls. In addition to cardiac, and aortic protection, ALL treatment for 10 or 12 weeks significantly reduced cerebrovascular damage incidence and scoring, along with a steady reduction of blood pressure (BP) in RHR-SP. Meanwhile, it significantly decreased serum aldosterone and malondialdehyde levels and cerebral NAD(P)H oxidase expressions in RHR-SP. We conducted 24 h continuous BP recording in conscious freely moving RHR-SP, and found that a single intragastric administration of ALL produced a long hypotensive effect lasting for at least 12 h on systolic BP. Taken together, our results in RHR-SP demonstrate that ALL can be used for stroke prevention via BP reduction and organ protection, with the molecular mechanisms related to inhibition of angiotensin-aldosterone system and oxidative stress. This study also provides a valuable scoring for evaluation of cerebrovascular damage and drug efficacy.
Collapse
Affiliation(s)
- Qi-Sheng Ling
- School of Pharmacy, Yantai University, Yantai, 264005, China
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Sai-Long Zhang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Jia-Sheng Tian
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Ming-He Cheng
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Ai-Jun Liu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Feng-Hua Fu
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Jian-Guo Liu
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
33
|
Sharma S, Advani D, Das A, Malhotra N, Khosla A, Arora V, Jha A, Yadav M, Ambasta RK, Kumar P. Pharmacological intervention in oxidative stress as a therapeutic target in neurological disorders. J Pharm Pharmacol 2021; 74:461-484. [PMID: 34050648 DOI: 10.1093/jpp/rgab064] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 04/01/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Oxidative stress is a major cellular burden that triggers reactive oxygen species (ROS) and antioxidants that modulate signalling mechanisms. Byproducts generated from this process govern the brain pathology and functions in various neurological diseases. As oxidative stress remains the key therapeutic target in neurological disease, it is necessary to explore the multiple routes that can significantly repair the damage caused due to ROS and consequently, neurodegenerative disorders (NDDs). Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase is the critical player of oxidative stress that can also be used as a therapeutic target to combat NDDs. KEY FINDINGS Several antioxidants signalling pathways are found to be associated with oxidative stress and show a protective effect against stressors by increasing the release of various cytoprotective enzymes and also exert anti-inflammatory response against this oxidative damage. These pathways along with antioxidants and reactive species can be the defined targets to eliminate or reduce the harmful effects of neurological diseases. SUMMARY Herein, we discussed the underlying mechanism and crucial role of antioxidants in therapeutics together with natural compounds as a pharmacological tool to combat the cellular deformities cascades caused due to oxidative stress.
Collapse
Affiliation(s)
- Sudhanshu Sharma
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Dia Advani
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Ankita Das
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Nishtha Malhotra
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Akanksha Khosla
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Vanshika Arora
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Ankita Jha
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Megha Yadav
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Department of Biotechnology, Delhi Technological University (Formerly DCE), Delhi, India
| |
Collapse
|
34
|
Kowluru RA. Diabetic Retinopathy and NADPH Oxidase-2: A Sweet Slippery Road. Antioxidants (Basel) 2021; 10:783. [PMID: 34063353 PMCID: PMC8156589 DOI: 10.3390/antiox10050783] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/03/2021] [Accepted: 05/13/2021] [Indexed: 11/17/2022] Open
Abstract
Diabetic retinopathy remains the leading cause of vision loss in working-age adults. The multi-factorial nature of the disease, along with the complex structure of the retina, have hindered in elucidating the exact molecular mechanism(s) of this blinding disease. Oxidative stress appears to play a significant role in its development and experimental models have shown that an increase in cytosolic Reacttive Oxygen Speies (ROS) due to the activation of NADPH oxidase 2 (Nox2), is an early event, which damages the mitochondria, accelerating loss of capillary cells. One of the integral proteins in the assembly of Nox2 holoenzyme, Rac1, is also activated in diabetes, and due to epigenetic modifications its gene transcripts are upregulated. Moreover, addition of hyperlipidemia in a hyperglycemic milieu (type 2 diabetes) further exacerbates Rac1-Nox2-ROS activation, and with time, this accelerates and worsens the mitochondrial damage, ultimately leading to the accelerated capillary cell loss and the development of diabetic retinopathy. Nox2, a multicomponent enzyme, is a good candidate to target for therapeutic interventions, and the inhibitors of Nox2 and Rac1 (and its regulators) are in experimental or clinical trials for other diseases; their possible use to prevent/halt retinopathy will be a welcoming sign for diabetic patients.
Collapse
Affiliation(s)
- Renu A Kowluru
- Department of Ophthalmology, Visual and Anatomical Sciences, Kresge Eye Institute, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
35
|
Nox2 Inhibition Regulates Stress Response and Mitigates Skeletal Muscle Fiber Atrophy during Simulated Microgravity. Int J Mol Sci 2021; 22:ijms22063252. [PMID: 33806917 PMCID: PMC8005132 DOI: 10.3390/ijms22063252] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/12/2021] [Accepted: 03/16/2021] [Indexed: 01/25/2023] Open
Abstract
Insufficient stress response and elevated oxidative stress can contribute to skeletal muscle atrophy during mechanical unloading (e.g., spaceflight and bedrest). Perturbations in heat shock proteins (e.g., HSP70), antioxidant enzymes, and sarcolemmal neuronal nitric oxidase synthase (nNOS) have been linked to unloading-induced atrophy. We recently discovered that the sarcolemmal NADPH oxidase-2 complex (Nox2) is elevated during unloading, downstream of angiotensin II receptor 1, and concomitant with atrophy. Here, we hypothesized that peptidyl inhibition of Nox2 would attenuate disruption of HSP70, MnSOD, and sarcolemmal nNOS during unloading, and thus muscle fiber atrophy. F344 rats were divided into control (CON), hindlimb unloaded (HU), and hindlimb unloaded +7.5 mg/kg/day gp91ds-tat (HUG) groups. Unloading-induced elevation of the Nox2 subunit p67phox-positive staining was mitigated by gp91ds-tat. HSP70 protein abundance was significantly lower in HU muscles, but not HUG. MnSOD decreased with unloading; however, MnSOD was not rescued by gp91ds-tat. In contrast, Nox2 inhibition protected against unloading suppression of the antioxidant transcription factor Nrf2. nNOS bioactivity was reduced by HU, an effect abrogated by Nox2 inhibition. Unloading-induced soleus fiber atrophy was significantly attenuated by gp91ds-tat. These data establish a causal role for Nox2 in unloading-induced muscle atrophy, linked to preservation of HSP70, Nrf2, and sarcolemmal nNOS.
Collapse
|
36
|
Fragoso-Morales LG, Correa-Basurto J, Rosales-Hernández MC. Implication of Nicotinamide Adenine Dinucleotide Phosphate (NADPH) Oxidase and Its Inhibitors in Alzheimer's Disease Murine Models. Antioxidants (Basel) 2021; 10:antiox10020218. [PMID: 33540840 PMCID: PMC7912941 DOI: 10.3390/antiox10020218] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/19/2021] [Accepted: 01/23/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is one of the main human dementias around the world which is constantly increasing every year due to several factors (age, genetics, environment, etc.) and there are no prevention or treatment options to cure it. AD is characterized by memory loss associated with oxidative stress (OS) in brain cells (neurons, astrocytes, microglia, etc.). OS can be produced by amyloid beta (Aβ) protein aggregation and its interaction with metals, mitochondrial damage and alterations between antioxidants and oxidant enzymes such as nicotinamide adenine dinucleotide phosphate (NADPH) oxidase. NADPH oxidase produces reactive oxygen species (ROS) and it is overexpressed in AD, producing large amounts of superoxide anions and hydrogen peroxide which damage brain cells and the vasculature. In addition, it has been reported that NADPH oxidase causes an imbalance of pH which could also influence in the amyloid beta (Aβ) production. Therefore, NADPH oxidase had been proposed as a therapeutic target in AD. However, there are no drugs for AD treatment such as an NADPH oxidase inhibitor despite great efforts made to stabilize the ROS production using antioxidant molecules. So, in this work, we will focus our attention on NADPH oxidase (NOX2 and NOX4) in AD as well as in AD models and later discuss the use of NADPH oxidase inhibitor compounds in AD.
Collapse
Affiliation(s)
- Leticia Guadalupe Fragoso-Morales
- Laboratorio de Biofísica y Biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Mexico City 11340, Mexico;
| | - José Correa-Basurto
- Laboratorio de Diseño y Desarrollo de Nuevos Fármacos e Innovación Biotecnológica, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón, Mexico City 11340, Mexico;
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatálisis, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Mexico City 11340, Mexico;
- Correspondence: ; Tel.: +(55)-572-960-00 (ext. 62767 & 62809)
| |
Collapse
|
37
|
Pratt SJP, Lee RM, Chang KT, Hernández-Ochoa EO, Annis DA, Ory EC, Thompson KN, Bailey PC, Mathias TJ, Ju JA, Vitolo MI, Schneider MF, Stains JP, Ward CW, Martin SS. Mechanoactivation of NOX2-generated ROS elicits persistent TRPM8 Ca 2+ signals that are inhibited by oncogenic KRas. Proc Natl Acad Sci U S A 2020; 117:26008-26019. [PMID: 33020304 PMCID: PMC7584994 DOI: 10.1073/pnas.2009495117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Changes in the mechanical microenvironment and mechanical signals are observed during tumor progression, malignant transformation, and metastasis. In this context, understanding the molecular details of mechanotransduction signaling may provide unique therapeutic targets. Here, we report that normal breast epithelial cells are mechanically sensitive, responding to transient mechanical stimuli through a two-part calcium signaling mechanism. We observed an immediate, robust rise in intracellular calcium (within seconds) followed by a persistent extracellular calcium influx (up to 30 min). This persistent calcium was sustained via microtubule-dependent mechanoactivation of NADPH oxidase 2 (NOX2)-generated reactive oxygen species (ROS), which acted on transient receptor potential cation channel subfamily M member 8 (TRPM8) channels to prolong calcium signaling. In contrast, the introduction of a constitutively active oncogenic KRas mutation inhibited the magnitude of initial calcium signaling and severely blunted persistent calcium influx. The identification that oncogenic KRas suppresses mechanically-induced calcium at the level of ROS provides a mechanism for how KRas could alter cell responses to tumor microenvironment mechanics and may reveal chemotherapeutic targets for cancer. Moreover, we find that expression changes in both NOX2 and TRPM8 mRNA predict poor clinical outcome in estrogen receptor (ER)-negative breast cancer patients, a population with limited available treatment options. The clinical and mechanistic data demonstrating disruption of this mechanically-activated calcium pathway in breast cancer patients and by KRas activation reveal signaling alterations that could influence cancer cell responses to the tumor mechanical microenvironment and impact patient survival.
Collapse
Affiliation(s)
- Stephen J P Pratt
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201;
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Rachel M Lee
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Katarina T Chang
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Erick O Hernández-Ochoa
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - David A Annis
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Eleanor C Ory
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Keyata N Thompson
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Patrick C Bailey
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Trevor J Mathias
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Julia A Ju
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Michele I Vitolo
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Martin F Schneider
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Joseph P Stains
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201
| | - Christopher W Ward
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD 21201
- School of Nursing, University of Maryland, Baltimore, MD 21201
| | - Stuart S Martin
- Department of Pharmacology, School of Medicine, University of Maryland, Baltimore, MD 21201;
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201
- Marlene and Stewart Greenebaum National Cancer Institute Comprehensive Cancer Center, School of Medicine, University of Maryland, Baltimore, MD 21201
| |
Collapse
|
38
|
An Iatrogenic Model of Brain Small-Vessel Disease: Post-Radiation Encephalopathy. Int J Mol Sci 2020; 21:ijms21186506. [PMID: 32899565 PMCID: PMC7555594 DOI: 10.3390/ijms21186506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 08/27/2020] [Accepted: 09/01/2020] [Indexed: 12/21/2022] Open
Abstract
We studied 114 primitive cerebral neoplasia, that were surgically treated, and underwent radiotherapy (RT), and compared their results to those obtained by 190 patients diagnosed with subcortical vascular dementia (sVAD). Patients with any form of primitive cerebral neoplasia underwent whole-brain radiotherapy. All the tumor patients had regional field partial brain RT, which encompassed each tumor, with an average margin of 2.6 cm from the initial target tumor volume. We observed in our patients who have been exposed to a higher dose of RT (30–65 Gy) a cognitive and behavior decline similar to that observed in sVAD, with the frontal dysexecutive syndrome, apathy, and gait alterations, but with a more rapid onset and with an overwhelming effect. Multiple mechanisms are likely to be involved in radiation-induced cognitive impairment. The active site of RT brain damage is the white matter areas, particularly the internal capsule, basal ganglia, caudate, hippocampus, and subventricular zone. In all cases, radiation damage inside the brain mainly focuses on the cortical–subcortical frontal loops, which integrate and process the flow of information from the cortical areas, where executive functions are “elaborated” and prepared, towards the thalamus, subthalamus, and cerebellum, where they are continuously refined and executed. The active mechanisms that RT drives are similar to those observed in cerebral small vessel disease (SVD), leading to sVAD. The RT’s primary targets, outside the tumor mass, are the blood–brain barrier (BBB), the small vessels, and putative mechanisms that can be taken into account are oxidative stress and neuro-inflammation, strongly associated with the alteration of NMDA receptor subunit composition.
Collapse
|
39
|
Marzaioli V, Canavan M, Floudas A, Wade SC, Low C, Veale DJ, Fearon U. Monocyte-Derived Dendritic Cell Differentiation in Inflammatory Arthritis Is Regulated by the JAK/STAT Axis via NADPH Oxidase Regulation. Front Immunol 2020; 11:1406. [PMID: 32733468 PMCID: PMC7358435 DOI: 10.3389/fimmu.2020.01406] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 06/01/2020] [Indexed: 01/15/2023] Open
Abstract
Monocyte-derived Dendritic cells (Mo-DC) are a distinct DC subset, involved in inflammation and infection, they originate from monocytes upon stimulation in the circulation and their activation and function may vary in autoimmune diseases. In this study we investigate the differences in Mo-DC differentiation and function in patients with Rheumatoid (RA) compared to Psoriatic arthritis (PsA). A significant increase in the Mo-DC differentiation marker CD209, paralleled by a corresponding decrease in the monocytic marker CD14, was demonstrated in RA compared to PsA, as early as 1 day post Mo-DC differentiation. RA monocytes ex-vivo were phenotypically different to PsA, displaying a more mature phenotype associated with altered cellular-morphology, early dendrite formation, and a significant increase in the CD40 marker. In addition, SPICE algorithm flow cytometric analysis showed distinct differences in chemokine receptors distribution in HC compared to PsA and RA CD14+ cells in the blood, with increased expression of the chemokine receptors CCR7 and CXCR4 observed in PsA and RA. In addition CD14+ cells at the site of inflammation showed a different chemokine receptor pattern between PsA and RA patients, with higher expression of CXCR3 and CXCR5 in RA when compared to PsA. The early priming observed in RA resulted in monocyte-endocytosis and antigen-uptake mechanisms to be impaired, effects that were not observed in PsA where phagocytosis capacity remained highly functional. Tofacitinib inhibited early Mo-DC differentiation, decreasing both CD209 and CD40 activation markers in RA. Inhibition of Mo-DC differentiation in response to Tofacitinib was mediated via an imbalance in the activation of NADPH-oxidases NOX5 and NOX2. This effect was reversed by NOX5 inhibition, but not NOX2, resulting in suppression of NOX5-dependent ROS production. In conclusion, RA monocytes are already primed ex vivo to become DC, evident by increased expression of activation markers, morphological appearance and impaired endocytosis capacity. Furthermore, we demonstrated for the first time that NOX5 mediates Mo-DC differentiation and function in response to Tofacitinib, which may alter DC functions.
Collapse
Affiliation(s)
- Viviana Marzaioli
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Mary Canavan
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Achilleas Floudas
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan C. Wade
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Candice Low
- Rheumatology EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Douglas J. Veale
- Rheumatology EULAR Centre of Excellence, Centre for Arthritis & Rheumatic Diseases, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Ursula Fearon
- Molecular Rheumatology, School of Medicine, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
40
|
Cao L, Li X, Xu R, Yao K, Yang W, Zhu H, Wang G, Zhang J. DUOX2, a common modulator in preventive effects of monoamine-based antidepressants on water immersion restraint stress- and indomethacin- induced gastric mucosal damage. Eur J Pharmacol 2020; 876:173058. [PMID: 32131022 DOI: 10.1016/j.ejphar.2020.173058] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/28/2020] [Accepted: 02/28/2020] [Indexed: 01/06/2023]
Abstract
Multiple kinds of monoamine-based antidepressants have been shown prophylactic effects in experimentally induced gastric ulcer. The loss of redox homeostasis plays a principle role in the development of peptic mucosal damage. Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases are one of the most important sources of reactive oxygen species within the gastrointestinal tract. It is unclear whether there are some common NADPH oxidases modulated by monoamine-based antidepressants in different gastric mucosal damage models. We explored the effects of selective serotonin-norepinephrine reuptake inhibitor (SNRI) duloxetine on the reactive oxygen species production and antioxidant capacity in the gastric mucosa of water immersion restraint (WIRS) or indomethacin treated rats, and examined the role of NADPH oxidases in the protective effects. Pretreated duloxetine prevented the increase of gastric mucosal NADPH oxidase activity and NADPH oxidase inhibitor apocynin dose-dependently protected gastric mucosa from damage by the two factors. Furthermore, dual oxidase 2 (DUOX2) and NADPH oxidase4 (NOX4) are involved in the protective effects of duloxetine in both models. We then examined NADPH oxidases expression modulated by the other monoamine-based antidepressants including selective serotonin reuptake inhibitor (SSRIs) fluoxetine, tricyclic agent (TCAs) amitriptyline and monoamine oxidase inhibitor (MAOs) moclobemide in the two models, and all the three antidepressants reduced the DUOX2 expression in the gastric mucosa. So DUOX2 was a common modulator in the preventive effects of all the monoamine-based antidepressants on WIRS- and indomethacin-induced gastric lesion. Our work provided a peripheral joint molecular target for monoamine modulatory antidepressants, which may be helpful to reveal the mechanisms of this kind of drugs more than monoamine regulation.
Collapse
Affiliation(s)
- Linyu Cao
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China
| | - Xulin Li
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China
| | - Ruiming Xu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China
| | - Kaiyun Yao
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China
| | - Wanqi Yang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China
| | - Hongliang Zhu
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China
| | - Guibin Wang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China.
| | - Jianjun Zhang
- Department of Pharmacology, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xian Nong Tan Street, Beijing, 100050, China.
| |
Collapse
|
41
|
Schröder K. NADPH oxidases: Current aspects and tools. Redox Biol 2020; 34:101512. [PMID: 32480354 PMCID: PMC7262010 DOI: 10.1016/j.redox.2020.101512] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/11/2020] [Accepted: 03/16/2020] [Indexed: 12/27/2022] Open
Abstract
Reactive oxygen species (ROS) have been shown or at least suggested to play an essential role for cellular signaling as second messengers. NADPH oxidases represent a source of controlled ROS formation. Accordingly, understanding the role of individual NADPH oxidases bears potential to interfere with intracellular signaling cascades without disturbing the signaling itself. Many tools have been developed to study or inhibit the functions and roles of the NADPH oxidases. This short review summarizes diseases, potentially associated with NADPH oxidases, genetically modified animals, and inhibitors.
Collapse
Affiliation(s)
- Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Fachbereich Medizin der Goethe-Universität, Theodor-Stern Kai 7, 60590, Frankfurt, Germany. https://
| |
Collapse
|
42
|
Nocella C, Cammisotto V, Bartimoccia S, Castellani V, Loffredo L, Pastori D, Pignatelli P, Sanguigni V, Violi F, Carnevale R. A novel role of MMP2 in regulating platelet NOX2 activation. Free Radic Biol Med 2020; 152:355-362. [PMID: 32268176 DOI: 10.1016/j.freeradbiomed.2020.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/13/2022]
Abstract
NOX2 has a key role for cellular production of reactive oxidant species (ROS) and although the mechanism of its activation is well known, little is known about its regulation. Metallo-proteinases (MMPs) regulate numerous protein activities both in physiological and pathological conditions but their interplay with NOX2 and ROS formation is still unclear. We performed experimental studies in human platelets and polymorphonuclear leukocytes (PMNs) to investigate the interplay of MMP2 with NOX2 activity. In collagen-stimulated platelets and in PMA-stimulated PMNs from healthy subjects, an immediate burst of ROS was detected at 10 min to then decline at 20 min. Coincidentally, sNOX2-dp, a split-off product of NOX2, increased and peaked at 10 min. ROS production was persistent whereas sNOX2dp is not released in cells treated with MMP2 inhibitor compared to other MMPs inhibitors. Western blot analysis showed the highest MMP2 expression on the cell membrane 10 min after stimulation. Moreover, the co-immunoprecipitation assay confirms the interaction between MMP2 and NOX2 that formed an active immuno-complex. Treating cells with NOX2ds-tat, an inhibitor of NADPH oxidase, significantly reduced ROS formation, sNOX2-dp, MMP2 expression and MMP2-NOX2-complex, which were all restored if cells were added with H2O2. The study provides the first evidence that MMP2 has a key role in blunting platelet NOX2 activity and eventually ROS formation.
Collapse
Affiliation(s)
- Cristina Nocella
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Italy
| | - Vittoria Cammisotto
- Department of General Surgery and Surgical Speciality Paride Stefanini, Sapienza University of Rome, 00161, Rome, Italy
| | - Simona Bartimoccia
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Italy
| | - Valentina Castellani
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Italy
| | - Lorenzo Loffredo
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Italy
| | - Daniele Pastori
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Italy
| | - Pasquale Pignatelli
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Italy; Mediterranea, Cardiocentro, 80122, Napoli, Italy
| | - Valerio Sanguigni
- Department of Internal Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Francesco Violi
- Department of Clinical Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Italy; Mediterranea, Cardiocentro, 80122, Napoli, Italy
| | - Roberto Carnevale
- Mediterranea, Cardiocentro, 80122, Napoli, Italy; Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100, Latina, Italy.
| |
Collapse
|
43
|
Maqbool A, Watt NT, Haywood N, Viswambharan H, Skromna A, Makava N, Visnagri A, Shawer HM, Bridge K, Muminov SK, Griffin K, Beech DJ, Wheatcroft SB, Porter KE, Simmons KJ, Sukumar P, Shah AM, Cubbon RM, Kearney MT, Yuldasheva NY. Divergent effects of genetic and pharmacological inhibition of Nox2 NADPH oxidase on insulin resistance-related vascular damage. Am J Physiol Cell Physiol 2020; 319:C64-C74. [PMID: 32401607 PMCID: PMC7468885 DOI: 10.1152/ajpcell.00389.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Insulin resistance leads to excessive endothelial cell (EC) superoxide generation and accelerated atherosclerosis. The principal source of superoxide from the insulin-resistant endothelium is the Nox2 isoform of NADPH oxidase. Here we examine the therapeutic potential of Nox2 inhibition on superoxide generation in saphenous vein ECs (SVECs) from patients with advanced atherosclerosis and type 2 diabetes and on vascular function, vascular damage, and lipid deposition in apolipoprotein E-deficient (ApoE−/−) mice with EC-specific insulin resistance (ESMIRO). To examine the effect of genetic inhibition of Nox2, ESMIRO mice deficient in ApoE−/− and Nox2 (ESMIRO/ApoE−/−/Nox2−/y) were generated and compared with ESMIRO/ApoE−/−/Nox2+/y littermates. To examine the effect of pharmacological inhibition of Nox2, we administered gp91dstat or scrambled peptide to ESMIRO/ApoE−/− mice. SVECs from diabetic patients had increased expression of Nox2 protein with concomitant increase in superoxide generation, which could be reduced by the Nox2 inhibitor gp91dstat. After 12 wk Western diet, ESMIRO/ApoE−/−/Nox2−/y mice had reduced EC superoxide generation and greater aortic relaxation to acetylcholine. ESMIRO/ApoE−/−/Nox2−/y mice developed more lipid deposition in the thoraco-abdominal aorta with multiple foci of elastin fragmentation at the level of the aortic sinus and greater expression of intercellular adhesion molecule-1 (ICAM-1). Gp91dstat reduced EC superoxide and lipid deposition in the thoraco-abdominal aorta of ESMIRO/ApoE−/− mice without causing elastin fragmentation or increased ICAM-1 expression. These results demonstrate that insulin resistance is characterized by increased Nox2-derived vascular superoxide. Complete deletion of Nox2 in mice with EC insulin resistance exacerbates, whereas partial pharmacological Nox2 inhibition protects against, insulin resistance-induced vascular damage.
Collapse
Affiliation(s)
- Azhar Maqbool
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Nicole T Watt
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Natalie Haywood
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Hema Viswambharan
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Anna Skromna
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Natalia Makava
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Asjad Visnagri
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Heba M Shawer
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Katherine Bridge
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | | | - Kathryn Griffin
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - David J Beech
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Stephen B Wheatcroft
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Karen E Porter
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Katie J Simmons
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Piruthivi Sukumar
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Ajay M Shah
- British Heart Foundation, Centre of Research Excellence, King's College London, London, United Kingdom
| | - Richard M Cubbon
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Mark T Kearney
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| | - Nadira Y Yuldasheva
- Leeds Institute for Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
44
|
Erlich JR, To EE, Liong S, Brooks R, Vlahos R, O'Leary JJ, Brooks DA, Selemidis S. Targeting Evolutionary Conserved Oxidative Stress and Immunometabolic Pathways for the Treatment of Respiratory Infectious Diseases. Antioxid Redox Signal 2020; 32:993-1013. [PMID: 32008371 PMCID: PMC7426980 DOI: 10.1089/ars.2020.8028] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Up until recently, metabolism has scarcely been referenced in terms of immunology. However, emerging evidence has shown that immune cells undergo an adaptation of metabolic processes, known as the metabolic switch. This switch is key to the activation, and sustained inflammatory phenotype in immune cells, which includes the production of cytokines and reactive oxygen species (ROS) that underpin infectious diseases, respiratory and cardiovascular disease, neurodegenerative disease, as well as cancer. Recent Advances: There is a burgeoning body of evidence that immunometabolism and redox biology drive infectious diseases. For example, influenza A virus (IAV) utilizes endogenous ROS production via NADPH oxidase (NOX)2-containing NOXs and mitochondria to circumvent antiviral responses. These evolutionary conserved processes are promoted by glycolysis, the pentose phosphate pathway, and the tricarboxylic acid (TCA) cycle that drive inflammation. Such metabolic products involve succinate, which stimulates inflammation through ROS-dependent stabilization of hypoxia-inducible factor-1α, promoting interleukin-1β production by the inflammasome. In addition, itaconate has recently gained significant attention for its role as an anti-inflammatory and antioxidant metabolite of the TCA cycle. Critical Issues: The molecular mechanisms by which immunometabolism and ROS promote viral and bacterial pathology are largely unknown. This review will provide an overview of the current paradigms with an emphasis on the roles of immunometabolism and ROS in the context of IAV infection and secondary complications due to bacterial infection such as Streptococcus pneumoniae. Future Directions: Molecular targets based on metabolic cell processes and ROS generation may provide novel and effective therapeutic strategies for IAV and associated bacterial superinfections.
Collapse
Affiliation(s)
- Jonathan R. Erlich
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Eunice E. To
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Stella Liong
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - Robert Brooks
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Ross Vlahos
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
| | - John J. O'Leary
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
- Department of Histopathology, Trinity College Dublin, Dublin, Ireland
- Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin, Ireland
| | - Doug A. Brooks
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
- Molecular Pathology Laboratory, Coombe Women and Infants' University Hospital, Dublin, Ireland
| | - Stavros Selemidis
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, Australia
- Address correspondence to: Prof. Stavros Selemidis, Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Bundoora, VIC 3083, Australia
| |
Collapse
|
45
|
To EE, Erlich JR, Liong F, Luong R, Liong S, Esaq F, Oseghale O, Anthony D, McQualter J, Bozinovski S, Vlahos R, O'Leary JJ, Brooks DA, Selemidis S. Mitochondrial Reactive Oxygen Species Contribute to Pathological Inflammation During Influenza A Virus Infection in Mice. Antioxid Redox Signal 2020; 32:929-942. [PMID: 31190565 PMCID: PMC7104903 DOI: 10.1089/ars.2019.7727] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Aims: Reactive oxygen species (ROS) are highly reactive molecules generated in different subcellular sites or compartments, including endosomes via the NOX2-containing nicotinamide adenine dinucleotide phosphate oxidase during an immune response and in mitochondria during cellular respiration. However, while endosomal NOX2 oxidase promotes innate inflammation to influenza A virus (IAV) infection, the role of mitochondrial ROS (mtROS) has not been comprehensively investigated in the context of viral infections in vivo. Results: In this study, we show that pharmacological inhibition of mtROS, with intranasal delivery of MitoTEMPO, resulted in a reduction in airway/lung inflammation, neutrophil infiltration, viral titers, as well as overall morbidity and mortality in mice infected with IAV (Hkx31, H3N2). MitoTEMPO treatment also attenuated apoptotic and necrotic neutrophils and macrophages in airway and lung tissue. At an early phase of influenza infection, that is, day 3 there were significantly lower amounts of IL-1β protein in the airways, but substantially higher amounts of type I IFN-β following MitoTEMPO treatment. Importantly, blocking mtROS did not appear to alter the initiation of an adaptive immune response by lung dendritic cells, nor did it affect lung B and T cell populations that participate in humoral and cellular immunity. Innovation/Conclusion: Influenza virus infection promotes mtROS production, which drives innate immune inflammation and this exacerbates viral pathogenesis. This pathogenic cascade highlights the therapeutic potential of local mtROS antioxidant delivery to alleviate influenza virus pathology.
Collapse
Affiliation(s)
- Eunice E To
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Jonathan R Erlich
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Felicia Liong
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Raymond Luong
- Infection and Immunity Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Melbourne, Australia
| | - Stella Liong
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Farisha Esaq
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Osezua Oseghale
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Desiree Anthony
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Jonathan McQualter
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Steven Bozinovski
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Ross Vlahos
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - John J O'Leary
- Department of Histopathology Trinity College Dublin, Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin, Ireland.,Molecular Pathology Laboratory, Coombe Women and Infants' University Hospital, Dublin, Ireland
| | - Doug A Brooks
- Division of Health Sciences, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, Australia
| | - Stavros Selemidis
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health Sciences and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| |
Collapse
|
46
|
Beyond bacterial killing: NADPH oxidase 2 is an immunomodulator. Immunol Lett 2020; 221:39-48. [DOI: 10.1016/j.imlet.2020.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 02/09/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
|
47
|
To EE, O'Leary JJ, O'Neill LAJ, Vlahos R, Bozinovski S, Porter CJH, Brooks RD, Brooks DA, Selemidis S. Spatial Properties of Reactive Oxygen Species Govern Pathogen-Specific Immune System Responses. Antioxid Redox Signal 2020; 32:982-992. [PMID: 32008365 PMCID: PMC7426979 DOI: 10.1089/ars.2020.8027] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significance: Reactive oxygen species (ROS) are often considered to be undesirable toxic molecules that are generated under conditions of cellular stress, which can cause damage to critical macromolecules such as DNA. However, ROS can also contribute to the pathogenesis of cancer and many other chronic inflammatory disease conditions, including atherosclerosis, metabolic disease, chronic obstructive pulmonary disease, neurodegenerative disease, and autoimmune disease. Recent Advances: The field of ROS biology is expanding, with an emerging paradigm that these reactive species are not generated haphazardly, but instead produced in localized regions or in specific subcellular compartments, and this has important consequences for immune system function. Currently, there is evidence for ROS generation in extracellular spaces, in endosomal compartments, and within mitochondria. Intriguingly, the specific location of ROS production appears to be influenced by the type of invading pathogen (i.e., bacteria, virus, or fungus), the size of the invading pathogen, as well as the expression/subcellular action of pattern recognition receptors and their downstream signaling networks, which sense the presence of these invading pathogens. Critical Issues: ROS are deliberately generated by the immune system, using specific NADPH oxidases that are critically important for pathogen clearance. Professional phagocytic cells can sense a foreign bacterium, initiate phagocytosis, and then within the confines of the phagosome, deliver bursts of ROS to these pathogens. The importance of confining ROS to this specific location is the impetus for this perspective. Future Directions: There are specific knowledge gaps on the fate of the ROS generated by NADPH oxidases/mitochondria, how these ROS are confined to specific locations, as well as the identity of ROS-sensitive targets and how they regulate cellular signaling.
Collapse
Affiliation(s)
- Eunice E To
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia.,Infection and Immunity Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - John J O'Leary
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland.,Sir Patrick Dun's Laboratory, Central Pathology Laboratory, St James's Hospital, Dublin, Ireland.,Emer Casey Research Laboratory, Molecular Pathology Laboratory, The Coombe Women and Infants University Hospital, Dublin, Ireland.,CERVIVA Research Consortium, Trinity College Dublin, Dublin, Ireland
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland
| | - Ross Vlahos
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Steven Bozinovski
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia
| | - Christopher J H Porter
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.,Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia
| | - Robert D Brooks
- School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Doug A Brooks
- Discipline of Histopathology, School of Medicine, Trinity Translational Medicine Institute (TTMI), Trinity College Dublin, Dublin, Ireland.,School of Pharmacy and Medical Sciences, University of South Australia Cancer Research Institute, University of South Australia, Adelaide, Australia
| | - Stavros Selemidis
- Program in Chronic Infectious and Inflammatory Diseases, Oxidant and Inflammation Biology Group, School of Health and Biomedical Sciences, College of Science, Engineering & Health, RMIT University, Melbourne, Australia.,Infection and Immunity Program, Department of Pharmacology, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| |
Collapse
|
48
|
Delgado NTB, Rouver WN, Dos Santos RL. Protective Effects of Pomegranate in Endothelial Dysfunction. Curr Pharm Des 2020; 26:3684-3699. [PMID: 32250215 DOI: 10.2174/1381612826666200406152147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/10/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Punica granatum L. is an infructescence native of occidental Asia and Mediterranean Europe, popularly referred to as pomegranate. It has been used in ethnomedicine for several applications, including the treatment of obesity, inflammation, diabetes, and the regulation of blood lipid parameters. Thus, pomegranate has been linked to the treatment of cardiovascular diseases that have endothelial dysfunction as a common factor acting mainly against oxidative stress due to its high polyphenol content. Its biocomponents have antihypertensive, antiatherogenic, antihyperglycemic, and anti-inflammatory properties, which promote cardiovascular protection through the improvement of endothelial function. METHODS Different electronic databases were searched in a non-systematic way to uncover the literature of interest. CONCLUSION This review article presents updated information on the role of pomegranate in the context of endothelial dysfunction and cardiovascular diseases. We have shown that pomegranate, or rather its components (e.g., tannins, flavonoids, phytoestrogens, anthocyanins, alkaloids, etc.), have beneficial effects on the cardiovascular system, improving parameters such as oxidative stress and the enzymatic antioxidant system, reducing reactive oxygen species formation and acting in an anti-inflammatory way. Thus, this review may contribute to a better understanding of pomegranate's beneficial actions on endothelial function and possibly to the development of strategies associated with conventional treatments of cardiovascular diseases.
Collapse
Affiliation(s)
- Nathalie T B Delgado
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - Wender N Rouver
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| | - Roger L Dos Santos
- Department of Physiological Sciences, Federal University of Espirito Santo, Vitoria, Brazil
| |
Collapse
|
49
|
Vilchis-Landeros M, Guinzberg R, Riveros-Rosas H, Villalobos-Molina R, Piña E. Aquaporin 8 is involved in H 2 O 2 -mediated differential regulation of metabolic signaling by α 1 - and β-adrenoceptors in hepatocytes. FEBS Lett 2020; 594:1564-1576. [PMID: 32115689 DOI: 10.1002/1873-3468.13763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species participate in regulating intracellular signaling pathways. Herein, we investigated the reported opposite effects of hydrogen peroxide (H2 O2 ) on metabolic signaling mediated by activated α1 - and β-adrenoceptors (ARs) in hepatocytes. In isolated rat hepatocytes, stimulation of α1 -AR increases H2 O2 production via NADPH oxidase 2 (NOX2) activation. We find that the H2 O2 thus produced is essential for α1 -AR-mediated activation of the classical hepatic glycogenolytic, gluconeogenic, and ureagenic responses. However, H2 O2 inhibits β-AR-mediated activation of these metabolic responses. We show that H2 O2 mediates its effects on α1 -AR and β-AR by permeating cells through aquaporin 8 (AQP8) channels and promoting Ca2+ mobilization. Thus, our findings reveal a novel NOX2-H2 O2 -AQP8-Ca2+ signaling cascade acting downstream of α1 -AR in hepatocytes, which, by negatively regulating β-AR signaling, establishes negative crosstalk between the two pathways.
Collapse
Affiliation(s)
- Magdalena Vilchis-Landeros
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Raquel Guinzberg
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Héctor Riveros-Rosas
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Rafael Villalobos-Molina
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México.,Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, México
| | - Enrique Piña
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
50
|
Obradovic M, Essack M, Zafirovic S, Sudar‐Milovanovic E, Bajic VP, Van Neste C, Trpkovic A, Stanimirovic J, Bajic VB, Isenovic ER. Redox control of vascular biology. Biofactors 2020; 46:246-262. [PMID: 31483915 PMCID: PMC7187163 DOI: 10.1002/biof.1559] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 08/14/2019] [Indexed: 12/12/2022]
Abstract
Redox control is lost when the antioxidant defense system cannot remove abnormally high concentrations of signaling molecules, such as reactive oxygen species (ROS). Chronically elevated levels of ROS cause oxidative stress that may eventually lead to cancer and cardiovascular and neurodegenerative diseases. In this review, we focus on redox effects in the vascular system. We pay close attention to the subcompartments of the vascular system (endothelium, smooth muscle cell layer) and give an overview of how redox changes influence those different compartments. We also review the core aspects of redox biology, cardiovascular physiology, and pathophysiology. Moreover, the topic-specific knowledgebase DES-RedoxVasc was used to develop two case studies, one focused on endothelial cells and the other on the vascular smooth muscle cells, as a starting point to possibly extend our knowledge of redox control in vascular biology.
Collapse
Affiliation(s)
- Milan Obradovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Magbubah Essack
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Sonja Zafirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Emina Sudar‐Milovanovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladan P. Bajic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Christophe Van Neste
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Andreja Trpkovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Julijana Stanimirovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| | - Vladimir B. Bajic
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE)ThuwalKingdom of Saudi Arabia
| | - Esma R. Isenovic
- Laboratory of Radiobiology and Molecular GeneticsVinca Institute of Nuclear Sciences, University of BelgradeBelgradeSerbia
| |
Collapse
|