1
|
Kim J, Moon JS. Molecular Roles of NADPH Oxidase-Mediated Oxidative Stress in Alzheimer's Disease: Isoform-Specific Contributions. Int J Mol Sci 2024; 25:12299. [PMID: 39596364 PMCID: PMC11594809 DOI: 10.3390/ijms252212299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Oxidative stress is linked to the pathogenesis of Alzheimer's disease (AD), a neurodegenerative disorder marked by memory impairment and cognitive decline. AD is characterized by the accumulation of amyloid-beta (Aβ) plaques and the formation of neurofibrillary tangles (NFTs) of hyperphosphorylated tau. AD is associated with an imbalance in redox states and excessive reactive oxygen species (ROS). Recent studies report that NADPH oxidase (NOX) enzymes are significant contributors to ROS generation in neurodegenerative diseases, including AD. NOX-derived ROS aggravates oxidative stress and neuroinflammation during AD. In this review, we provide the potential role of all NOX isoforms in AD pathogenesis and their respective structural involvement in AD progression, highlighting NOX enzymes as a strategic therapeutic target. A comprehensive understanding of NOX isoforms and their inhibitors could provide valuable insights into AD pathology and aid in the development of targeted treatments for AD.
Collapse
Affiliation(s)
- Junhyung Kim
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea;
| | - Jong-Seok Moon
- Department of Integrated Biomedical Science, Soonchunhyang Institute of Medi-Bio Science (SIMS), Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea;
- Department of Pathology, College of Medicine, Soonchunhyang University, Cheonan 31151, Chungcheongnam-do, Republic of Korea
| |
Collapse
|
2
|
Seo YS, Park KH, Park JM, Jeong H, Kim B, Jeon JS, Yu J, Kim SK, Lee K, Lee MY. Short-term inhalation exposure to cigarette smoke induces oxidative stress and inflammation in lungs without systemic oxidative stress in mice. Toxicol Res 2024; 40:273-283. [PMID: 38525133 PMCID: PMC10959912 DOI: 10.1007/s43188-023-00223-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 12/23/2023] [Accepted: 12/27/2023] [Indexed: 03/26/2024] Open
Abstract
Smoking is a well-established risk factor for various pathologies, including pulmonary diseases, cardiovascular disorders, and cancers. The toxic effects of cigarette smoke (CS) are mediated through multiple pathways and diverse mechanisms. A key pathogenic factor is oxidative stress, primarily induced by excessive formation of reactive oxygen species. However, it remains unclear whether smoking directly induces systemic oxidative stress or if such stress is a secondary consequence. This study aimed to determine whether short-term inhalation exposure to CS induces oxidative stress in extrapulmonary organs in addition to the lung in a murine model. In the experiment, 3R4F reference cigarettes were used to generate CS, and 8-week-old male BALB/c mice were exposed to CS at a total particulate matter concentration of either 0 or 800 µg/L for four consecutive days. CS exposure led to an increase in neutrophils, eosinophils, and total cell counts in bronchoalveolar lavage fluid. It also elevated levels of lactate dehydrogenase and malondialdehyde (MDA), markers indicative of tissue damage and oxidative stress, respectively. Conversely, no significant changes were observed in systemic oxidative stress markers such as total oxidant scavenging capacity, MDA, glutathione (GSH), and the GSH/GSSG ratio in blood samples. In line with these findings, CS exposure elevated NADPH oxidase (NOX)-dependent superoxide generation in the lung but not in other organs like the liver, kidney, heart, aorta, and brain. Collectively, our results indicate that short-term exposure to CS induces inflammation and oxidative stress in the lung without significantly affecting oxidative stress in extrapulmonary organs under the current experimental conditions. NOX may play a role in these pulmonary-specific events.
Collapse
Affiliation(s)
- Yoon-Seok Seo
- College of Pharmacy, BK21 FOUR Team and Integrated Research Institute for Drug Development, Dongguk University, Goyang-si, Gyeonggi-do 10326 Republic of Korea
| | - Kwang-Hoon Park
- College of Pharmacy, BK21 FOUR Team and Integrated Research Institute for Drug Development, Dongguk University, Goyang-si, Gyeonggi-do 10326 Republic of Korea
| | - Jung-Min Park
- College of Pharmacy, BK21 FOUR Team and Integrated Research Institute for Drug Development, Dongguk University, Goyang-si, Gyeonggi-do 10326 Republic of Korea
| | - Hyuneui Jeong
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Jeollabuk-do 54596 Republic of Korea
| | - Bumseok Kim
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan-si, Jeollabuk-do 54596 Republic of Korea
| | - Jang Su Jeon
- College of Pharmacy, Chungnam National University, Daejeon, 34134 Republic of Korea
| | - Jieun Yu
- College of Pharmacy, Chungnam National University, Daejeon, 34134 Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon, 34134 Republic of Korea
| | - Kyuhong Lee
- Inhalation Toxicology Center for Airborne Risk Factor, Korea Institute of Toxicology, Jeongeup-si, Jeollabuk-do 56212 Republic of Korea
| | - Moo-Yeol Lee
- College of Pharmacy, BK21 FOUR Team and Integrated Research Institute for Drug Development, Dongguk University, Goyang-si, Gyeonggi-do 10326 Republic of Korea
| |
Collapse
|
3
|
Herb M. NADPH Oxidase 3: Beyond the Inner Ear. Antioxidants (Basel) 2024; 13:219. [PMID: 38397817 PMCID: PMC10886416 DOI: 10.3390/antiox13020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Reactive oxygen species (ROS) were formerly known as mere byproducts of metabolism with damaging effects on cellular structures. The discovery and description of NADPH oxidases (Nox) as a whole enzyme family that only produce this harmful group of molecules was surprising. After intensive research, seven Nox isoforms were discovered, described and extensively studied. Among them, the NADPH oxidase 3 is the perhaps most underrated Nox isoform, since it was firstly discovered in the inner ear. This stigma of Nox3 as "being only expressed in the inner ear" was also used by me several times. Therefore, the question arose whether this sentence is still valid or even usable. To this end, this review solely focuses on Nox3 and summarizes its discovery, the structural components, the activating and regulating factors, the expression in cells, tissues and organs, as well as the beneficial and detrimental effects of Nox3-mediated ROS production on body functions. Furthermore, the involvement of Nox3-derived ROS in diseases progression and, accordingly, as a potential target for disease treatment, will be discussed.
Collapse
Affiliation(s)
- Marc Herb
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, 50935 Cologne, Germany;
- German Centre for Infection Research, Partner Site Bonn-Cologne, 50931 Cologne, Germany
- Cologne Cluster of Excellence on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| |
Collapse
|
4
|
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: 33] [Impact Index Per Article: 16.5] [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
|
5
|
Bang J, Son KH, Heo HR, Park E, Kwak HJ, Uhm KO, Chung MH, Kim YY, Lim HJ. Exogenous 8-Hydroxydeoxyguanosine Attenuates PM 2.5-Induced Inflammation in Human Bronchial Epithelial Cells by Decreasing NLRP3 Inflammasome Activation. Antioxidants (Basel) 2023; 12:1189. [PMID: 37371919 DOI: 10.3390/antiox12061189] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/20/2023] [Accepted: 05/28/2023] [Indexed: 06/29/2023] Open
Abstract
Particulate matter 2.5 (PM2.5) induces lung injury by increasing the generation of reactive oxygen species (ROS) and inflammation. ROS aggravates NLRP3 inflammasome activation, which activates caspase-1, IL-1β, and IL-18 and induces pyroptosis; these factors propagate inflammation. In contrast, treatment with exogenous 8-hydroxydeoxyguanosine (8-OHdG) decreases RAC1 activity and eventually decreases dinucleotide phosphate oxidase (NOX) and ROS generation. To establish modalities that would mitigate PM2.5-induced lung injury, we evaluated whether 8-OHdG decreased PM2.5-induced ROS generation and NLRP3 inflammasome activation in BEAS-2B cells. CCK-8 and lactate dehydrogenase assays were used to determine the treatment concentration. Fluorescence intensity, Western blotting, enzyme-linked immunosorbent assay, and immunoblotting assays were also performed. Treatment with 80 μg/mL PM2.5 increased ROS generation, RAC1 activity, NOX1 expression, NLRP3 inflammasome (NLRP3, ASC, and caspase-1) activity, and IL-1β and IL-18 levels in cells; treatment with 10 μg/mL 8-OHdG significantly attenuated these effects. Furthermore, similar results, such as reduced expression of NOX1, NLRP3, ASC, and caspase-1, were observed in PM2.5-treated BEAS-2B cells when treated with an RAC1 inhibitor. These results show that 8-OHdG mitigates ROS generation and NLRP3 inflammation by inhibiting RAC1 activity and NOX1 expression in respiratory cells exposed to PM2.5.
Collapse
Affiliation(s)
- Jihye Bang
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Kuk Hui Son
- Gachon University Gil Medical Center, Department of Thoracic and Cardiovascular Surgery, College of Medicine, Gachon University, 21, Namdong-daero 774 beon-gil, Namdong-gu, Incheon 21565, Republic of Korea
| | - Hye-Ryeon Heo
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Eunsook Park
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Hyun-Jeong Kwak
- Major of Life Science, Division of Bioconvergence, College of Convergence and Integrated Science, Kyonggi University, 154-42 Gwanggosan-ro, Yeongtong-gu, Suwon-si 16227, Republic of Korea
| | - Kyung-Ok Uhm
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Myung-Hee Chung
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, 155, Gaetbeol-ro, Yeonsu-ku, Incheon 21999, Republic of Korea
| | - Young-Youl Kim
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| | - Hyun Joung Lim
- Division of Allergy and Respiratory Disease Research, Department of Chronic Disease Convergence Research, National Institute of Health, Osong Health Technology Administration Complex 187, Osongsaengmyeong 2-ro, Osong-eup, Heungdeok-gu, Cheongju-si 28159, Republic of Korea
| |
Collapse
|
6
|
Haq S, Sarodaya N, Karapurkar JK, Suresh B, Jo JK, Singh V, Bae YS, Kim KS, Ramakrishna S. CYLD destabilizes NoxO1 protein by promoting ubiquitination and regulates prostate cancer progression. Cancer Lett 2022; 525:146-157. [PMID: 34742871 DOI: 10.1016/j.canlet.2021.10.032] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/05/2021] [Accepted: 10/18/2021] [Indexed: 01/03/2023]
Abstract
The NADPH oxidase (Nox) family of enzymes is solely dedicated in the generation of reactive oxygen species (ROS). ROS generated by Nox are involved in multiple signaling cascades and a myriad of pathophysiological conditions including cancer. As such, ROS seem to have both detrimental and beneficial roles in a number of cellular functions, including cell signaling, growth, apoptosis and proliferation. Regulatory mechanisms are required to control the activity of Nox enzymes in order to maintain ROS balance within the cell. Here, we performed genome-wide screening for deubiquitinating enzymes (DUBs) regulating Nox organizer 1 (NoxO1) protein expression using a CRISPR/Cas9-mediated DUB-knockout library. We identified cylindromatosis (CYLD) as a binding partner regulating NoxO1 protein expression. We demonstrated that the overexpression of CYLD promotes ubiquitination of NoxO1 protein and reduces the NoxO1 protein half-life. The destabilization of NoxO1 protein by CYLD suppressed excessive ROS generation. Additionally, CRISPR/Cas9-mediated knockout of CYLD in PC-3 cells promoted cell proliferation, migration, colony formation and invasion in vitro. In xenografted mice, injection of CYLD-depleted cells consistently led to tumor development with increased weight and volume. Taken together, these results indicate that CYLD acts as a destabilizer of NoxO1 protein and could be a potential tumor suppressor target for cancer therapeutics.
Collapse
Affiliation(s)
- Saba Haq
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul, 04763, South Korea
| | - Neha Sarodaya
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| | | | - Bharathi Suresh
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea
| | - Jung Ki Jo
- Department of Urology, Hanyang University College of Medicine, Seoul, 04763, South Korea
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, Gujarat, India
| | - Yun Soo Bae
- Department of Life Science, Ewha Womans University, Seoul, South Korea
| | - Kye-Seong Kim
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| | - Suresh Ramakrishna
- Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul 04763, South Korea; College of Medicine, Hanyang University, Seoul, 04763, South Korea.
| |
Collapse
|
7
|
Kervin TA, Wiseman BC, Overduin M. Phosphoinositide Recognition Sites Are Blocked by Metabolite Attachment. Front Cell Dev Biol 2021; 9:690461. [PMID: 34368138 PMCID: PMC8340361 DOI: 10.3389/fcell.2021.690461] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 06/18/2021] [Indexed: 12/16/2022] Open
Abstract
Membrane readers take part in trafficking and signaling processes by localizing proteins to organelle surfaces and transducing molecular information. They accomplish this by engaging phosphoinositides (PIs), a class of lipid molecules which are found in different proportions in various cellular membranes. The prototypes are the PX domains, which exhibit a range of specificities for PIs. Our meta-analysis indicates that recognition of membranes by PX domains is specifically controlled by modification of lysine and arginine residues including acetylation, hydroxyisobutyrylation, glycation, malonylation, methylation and succinylation of sidechains that normally bind headgroups of phospholipids including organelle-specific PI signals. Such metabolite-modulated residues in lipid binding elements are named MET-stops here to highlight their roles as erasers of membrane reader functions. These modifications are concentrated in the membrane binding sites of half of all 49 PX domains in the human proteome and correlate with phosphoregulatory sites, as mapped using the Membrane Optimal Docking Area (MODA) algorithm. As these motifs are mutated and modified in various cancers and the responsible enzymes serve as potential drug targets, the discovery of MET-stops as a widespread inhibitory mechanism may aid in the development of diagnostics and therapeutics aimed at the readers, writers and erasers of the PI code.
Collapse
Affiliation(s)
- Troy A Kervin
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Brittany C Wiseman
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.,Molecular and Cellular Biology, MacEwan University, Edmonton, AB, Canada.,SMALP Network, Edmonton, AB, Canada
| | - Michael Overduin
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada.,SMALP Network, Edmonton, AB, Canada
| |
Collapse
|
8
|
Sindona C, Schepici G, Contestabile V, Bramanti P, Mazzon E. NOX2 Activation in COVID-19: Possible Implications for Neurodegenerative Diseases. ACTA ACUST UNITED AC 2021; 57:medicina57060604. [PMID: 34208136 PMCID: PMC8230853 DOI: 10.3390/medicina57060604] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 06/04/2021] [Accepted: 06/09/2021] [Indexed: 12/11/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is a rapidly spreading contagious infectious disease caused by the pathogen severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that primarily affects the respiratory tract as well as the central nervous system (CNS). SARS-CoV-2 infection occurs through the interaction of the viral protein Spike with the angiotensin II receptor (ACE 2), leading to an increase of angiotensin II and activation of nicotinamide adenine dinucleotide phosphate oxidase2 (NOX2), resulting in the release of both reactive oxygen species (ROS) and inflammatory molecules. The purpose of the review is to explain that SARS-CoV-2 infection can determine neuroinflammation that induces NOX2 activation in microglia. To better understand the role of NOX2 in inflammation, an overview of its involvement in neurodegenerative diseases (NDs) such as Parkinson’s disease (PD), Alzheimer’s disease (AD), and amyotrophic lateral sclerosis (ALS) is provided. To write this manuscript, we performed a PubMed search to evaluate the possible relationship of SARS-CoV-2 infection in NOX2 activation in microglia, as well as the role of NOX2 in NDs. Several studies highlighted that NOX2 activation in microglia amplifies neuroinflammation. To date, there is no clinical treatment capable of counteracting its activation, however, NOX2 could be a promising pharmaceutical target useful for both the treatment and prevention of NDs and COVID-19 treatment.
Collapse
|
9
|
Kervin TA, Overduin M. Regulation of the Phosphoinositide Code by Phosphorylation of Membrane Readers. Cells 2021; 10:cells10051205. [PMID: 34069055 PMCID: PMC8156045 DOI: 10.3390/cells10051205] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Revised: 05/07/2021] [Accepted: 05/09/2021] [Indexed: 02/07/2023] Open
Abstract
The genetic code that dictates how nucleic acids are translated into proteins is well known, however, the code through which proteins recognize membranes remains mysterious. In eukaryotes, this code is mediated by hundreds of membrane readers that recognize unique phosphatidylinositol phosphates (PIPs), which demark organelles to initiate localized trafficking and signaling events. The only superfamily which specifically detects all seven PIPs are the Phox homology (PX) domains. Here, we reveal that throughout evolution, these readers are universally regulated by the phosphorylation of their PIP binding surfaces based on our analysis of existing and modelled protein structures and phosphoproteomic databases. These PIP-stops control the selective targeting of proteins to organelles and are shown to be key determinants of high-fidelity PIP recognition. The protein kinases responsible include prominent cancer targets, underscoring the critical role of regulated membrane readership.
Collapse
|
10
|
Asaoka N, Ibi M, Hatakama H, Nagaoka K, Iwata K, Matsumoto M, Katsuyama M, Kaneko S, Yabe-Nishimura C. NOX1/NADPH Oxidase Promotes Synaptic Facilitation Induced by Repeated D 2 Receptor Stimulation: Involvement in Behavioral Repetition. J Neurosci 2021; 41:2780-2794. [PMID: 33563722 PMCID: PMC8018731 DOI: 10.1523/jneurosci.2121-20.2021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/21/2021] [Accepted: 01/27/2021] [Indexed: 11/21/2022] Open
Abstract
Repetitive behavior is a widely observed neuropsychiatric symptom. Abnormal dopaminergic signaling in the striatum is one of the factors associated with behavioral repetition; however, the molecular mechanisms underlying the induction of repetitive behavior remain unclear. Here, we demonstrated that the NOX1 isoform of the superoxide-producing enzyme NADPH oxidase regulated repetitive behavior in mice by facilitating excitatory synaptic inputs in the central striatum (CS). In male C57Bl/6J mice, repeated stimulation of D2 receptors induced abnormal behavioral repetition and perseverative behavior. Nox1 deficiency or acute pharmacological inhibition of NOX1 significantly shortened repeated D2 receptor stimulation-induced repetitive behavior without affecting motor responses to a single D2 receptor stimulation. Among brain regions, Nox1 showed enriched expression in the striatum, and repeated dopamine D2 receptor stimulation further increased Nox1 expression levels in the CS, but not in the dorsal striatum. Electrophysiological analyses revealed that repeated D2 receptor stimulation facilitated excitatory inputs in the CS indirect pathway medium spiny neurons (iMSNs), and this effect was suppressed by the genetic deletion or pharmacological inhibition of NOX1. Nox1 deficiency potentiated protein tyrosine phosphatase activity and attenuated the accumulation of activated Src kinase, which is required for the synaptic potentiation in CS iMSNs. Inhibition of NOX1 or β-arrestin in the CS was sufficient to ameliorate repetitive behavior. Striatal-specific Nox1 knockdown also ameliorated repetitive and perseverative behavior. Collectively, these results indicate that NOX1 acts as an enhancer of synaptic facilitation in CS iMSNs and plays a key role in the molecular link between abnormal dopamine signaling and behavioral repetition and perseveration.SIGNIFICANCE STATEMENT Behavioral repetition is a form of compulsivity, which is one of the core symptoms of psychiatric disorders, such as obsessive-compulsive disorder. Perseveration is also a hallmark of such disorders. Both clinical and animal studies suggest important roles of abnormal dopaminergic signaling and striatal hyperactivity in compulsivity; however, the precise molecular link between them remains unclear. Here, we demonstrated the contribution of NOX1 to behavioral repetition induced by repeated stimulation of D2 receptors. Repeated stimulation of D2 receptors upregulated Nox1 mRNA in a striatal subregion-specific manner. The upregulated NOX1 promoted striatal synaptic facilitation in iMSNs by enhancing phosphorylation signaling. These results provide a novel mechanism for D2 receptor-mediated excitatory synaptic facilitation and indicate the therapeutic potential of NOX1 inhibition in compulsivity.
Collapse
Affiliation(s)
- Nozomi Asaoka
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masakazu Ibi
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Hikari Hatakama
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Koki Nagaoka
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Kazumi Iwata
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Misaki Matsumoto
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Masato Katsuyama
- Radioisotope Center, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Chihiro Yabe-Nishimura
- Department of Pharmacology, Kyoto Prefectural University of Medicine, Kyoto, 602-8566, Japan
| |
Collapse
|
11
|
Jäckle K, Zeis T, Schaeren-Wiemers N, Junker A, van der Meer F, Kramann N, Stadelmann C, Brück W. Molecular signature of slowly expanding lesions in progressive multiple sclerosis. Brain 2020; 143:2073-2088. [PMID: 32577755 DOI: 10.1093/brain/awaa158] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/04/2020] [Accepted: 03/30/2020] [Indexed: 01/08/2023] Open
Abstract
Multiple sclerosis is an immune-mediated chronic inflammatory disease of the CNS that leads to demyelinated lesions in the grey and white matter. Inflammatory, active demyelinating white matter lesions predominate in the relapsing-remitting disease stages, whereas in the progressive stage the so-called slowly expanding lesion is characteristic. These lesions show an accumulation of macrophages/microglia at their borders, mediating the ongoing myelin breakdown and axonal degeneration. The exact pathogenetic mechanisms of lesion progression in chronic multiple sclerosis are still not clear. In the present study, we performed a detailed immunological and molecular profiling of slowly expanding lesions (n = 21) from 13 patients aged between 30 to 74 years (five females and eight males), focusing on macrophage/microglia differentiation. By applying the microglia-specific marker TMEM119, we demonstrate that cells accumulating at the lesion edge almost exclusively belonged to the microglia lineage. Macrophages/microglia can be subdivided into the M1 type, which are associated with inflammatory and degenerative processes, and M2 type, with protective properties, whereby also intermediate polarization phenotypes can be observed. By using a panel of markers characterizing M1- or M2-type macrophages/microglia, we observed a preferential accumulation of M1-type differentiated cells at the lesion edge, indicating a crucial role of these cells in lesion progression. Additionally, unbiased RNA microarray analyses of macrodissected lesion edges from slowly expanding and chronic inactive lesions as well as normal-appearing white matter were performed. In slowly expanding lesions, we identified a total of 165 genes that were upregulated and 35 genes that were downregulated. The upregulated genes included macrophage/microglia-associated genes involved in immune defence and inflammatory processes. Among the upregulated genes were ALOX15B, MME and TNFRSF25. We confirmed increased expression of ALOX15B by quantitative PCR, and of all three genes on the protein level by immunohistochemistry. In conclusion, the present study characterized in detail slowly expanding lesions in progressive multiple sclerosis and demonstrated a preferential accumulation of resident microglia with M1 differentiation at the lesion edge. Microarray analysis showed an increased expression of genes related to immune function, metabolic processes as well as transcription/translation. Thus, these genes may serve as future therapeutic targets to impede lesion progression.
Collapse
Affiliation(s)
- Katharina Jäckle
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen, Göttingen, Germany
| | - Thomas Zeis
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Nicole Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Andreas Junker
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.,Department of Neuropathology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | | | - Nadine Kramann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolfgang Brück
- Institute of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
12
|
Buchmann GK, Schürmann C, Warwick T, Schulz MH, Spaeth M, Müller OJ, Schröder K, Jo H, Weissmann N, Brandes RP. Deletion of NoxO1 limits atherosclerosis development in female mice. Redox Biol 2020; 37:101713. [PMID: 32949971 PMCID: PMC7502371 DOI: 10.1016/j.redox.2020.101713] [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] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 08/28/2020] [Accepted: 08/29/2020] [Indexed: 01/22/2023] Open
Abstract
OBJECTIVE Oxidative stress is a risk factor for atherosclerosis. NADPH oxidases of the Nox family produce ROS but their contribution to atherosclerosis development is less clear. Nox2 promotes and Nox4 rather limits atherosclerosis. Although Nox1 with its cytosolic co-factors are largely expressed in epithelial cells, a role for Nox1 for atherosclerosis development was suggested. To further define the role of this homologue, the role of its essential cytosolic cofactor, NoxO1, was determined for atherosclerosis development with the aid of knockout mice. METHODS AND RESULTS Wildtype (WT) and NoxO1 knockout mice were treated with high fat diet and adeno-associated virus (AAV) overexpressing pro-protein convertase subtilisin/kexin type 9 (PCSK9) to induce hepatic low-density lipoprotein (LDL) receptor loss. As a result, massive hypercholesterolemia was induced and spontaneous atherosclerosis developed within three month. Deletion of NoxO1 reduced atherosclerosis formation in brachiocephalic artery and aortic arch in female but not male NoxO1-/- mice as compared to WT littermates. This was associated with a reduced pro-inflammatory cytokine signature in the plasma of female but not male NoxO1-/- mice. MACE-RNAseq of the vessel did not reveal this signature and the expression of the Nox1/NoxO1 system was low to not detectable. CONCLUSIONS The scaffolding protein NoxO1 plays some role in atherosclerosis development in female mice probably by attenuating the global inflammatory burden.
Collapse
Affiliation(s)
- Giulia K Buchmann
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany
| | - Tim Warwick
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany
| | - Marcel H Schulz
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany; Institute for Cardiovascular Regeneration, Goethe-University, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany
| | - Manuela Spaeth
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany
| | - Oliver J Müller
- Department of Internal Medicine III, University of Kiel, Arnold-Heller-Straße 3, 24105, Kiel, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Arnold-Heller-Straße 3, 24105, Kiel, Germany
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Ludwigstraße 23, 35390, Gießen, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe-University, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Theodor-Stern Kai 7, 60590, Frankfurt Am Main, Germany.
| |
Collapse
|
13
|
Miyano K, Okamoto S, Yamauchi A, Kajikawa M, Kiyohara T, Taura M, Kawai C, Kuribayashi F. Constitutive activity of NADPH oxidase 1 (Nox1) that promotes its own activity suppresses the colon epithelial cell migration. Free Radic Res 2020; 54:640-648. [PMID: 32924676 DOI: 10.1080/10715762.2020.1823383] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Superoxide producing NADPH oxidase 1 (Nox1), abundantly expressed in the colon epithelium, plays a crucial role in mucosal host defenses. In this study, we found that pre-treatment of cells with edaravone, a free radical scavenger, inhibited Nox1 constitutive activity even after washout without affecting Nox1 trafficking to the plasma membrane and membrane recruitment of the cytosolic regulators Noxo1 and Noxa1. These results suggest that a Nox1-derived product is involved in the step that initiates the electron transfer reaction after the formation of the Nox1-Noxo1-Noxa1 complex. Furthermore, we show that the mean migration directionality and velocity of epithelial cells were significantly enhanced by the inhibition of constitutive Nox1 activity. Thus, the constitutive Nox1 activity limits undesired cell migration in resting cells while participating in a positive feedback loop toward its own oxidase activity.
Collapse
Affiliation(s)
- Kei Miyano
- Department of Biochemistry, Kawasaki Medical School, Okayama, Japan
| | | | - Akira Yamauchi
- Department of Biochemistry, Kawasaki Medical School, Okayama, Japan
| | - Mizuho Kajikawa
- Laboratory of Microbiology, Showa Pharmaceutical University, Machida, Japan
| | - Takuya Kiyohara
- Department of Cerebrovascular Disease and Neurology, Hakujyuji Hospital, Fukuoka, Japan
| | - Masahiko Taura
- Department of Otorhinolaryngology, Faculty of medicine, Fukuoka University, Fukuoka, Japan
| | - Chikage Kawai
- Department of Biochemistry, Kawasaki Medical School, Okayama, Japan
| | | |
Collapse
|
14
|
Dang PMC, Rolas L, El-Benna J. The Dual Role of Reactive Oxygen Species-Generating Nicotinamide Adenine Dinucleotide Phosphate Oxidases in Gastrointestinal Inflammation and Therapeutic Perspectives. Antioxid Redox Signal 2020; 33:354-373. [PMID: 31968991 DOI: 10.1089/ars.2020.8018] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Despite their intrinsic cytotoxic properties, mounting evidence indicates that reactive oxygen species (ROS) physiologically produced by the nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (NOXs) of epithelial cells (NOX1, dual oxidase [DUOX]2) and phagocytes (NOX2) are critical for innate immune response and homeostasis of the intestinal mucosa. However, dysregulated ROS production could be a driving factor in inflammatory bowel diseases (IBDs). Recent Advances: In addition to NOX2, recent studies have demonstrated that NOX1- and DUOX2-derived ROS can regulate intestinal innate immune defense and homeostasis by impacting many processes, including bacterial virulence, expression of bacteriostatic proteins, epithelial renewal and restitution, and microbiota composition. Moreover, the antibacterial role of DUOX2 is a function conserved in evolution as it has been described in invertebrates, and lower and higher vertebrates. In humans, variants of the NOX2, NOX1, and DUOX2 genes, which are associated with impaired ROS production, have been identified in very early onset IBD, but overexpression of NOX/DUOX, especially DUOX2, has also been described in IBD, suggesting that loss-of-function or excessive activity of the ROS-generating enzymes could contribute to disease progression. Critical Issues: Therapeutic perspectives aiming at targeting NOX/DUOX in IBD should take into account the two sides of NOX/DUOX-derived ROS in intestinal inflammation. Hence, NOX/DUOX inhibitors or ROS inducers should be considered as a function of the disease context. Future Directions: A thorough understanding of the physiological and pathological regulation of NOX/DUOX in the gastrointestinal tract is an absolute pre-requisite for the development of therapeutic strategies that can modulate ROS levels in space and time.
Collapse
Affiliation(s)
- Pham My-Chan Dang
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Faculté de Médecine, Laboratoire d'Excellence Inflamex, DHU FIRE, Université de Paris, Paris, France
| | - Loïc Rolas
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France
| | - Jamel El-Benna
- INSERM-U1149, CNRS-ERL8252, Centre de Recherche sur l'Inflammation, Paris, France.,Faculté de Médecine, Laboratoire d'Excellence Inflamex, DHU FIRE, Université de Paris, Paris, France
| |
Collapse
|
15
|
Laddha AP, Kulkarni YA. NADPH oxidase: A membrane-bound enzyme and its inhibitors in diabetic complications. Eur J Pharmacol 2020; 881:173206. [PMID: 32442539 DOI: 10.1016/j.ejphar.2020.173206] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/08/2020] [Accepted: 05/14/2020] [Indexed: 12/20/2022]
Abstract
The human body has a mechanism for balancing the generation and neutralization of reactive oxygen species. The body is exposed to many agents that are responsible for the generation of reactive oxygen/nitrogen species, which leads to disruption of the balance between generation of these species and oxidative stress defence mechanisms. Diabetes is a chronic pathological condition associated with prolonged hyperglycaemia. Prolonged elevation of level of glucose in the blood leads to the generation of reactive oxygen species. This generation of reactive oxygen species is responsible for the development of diabetic vasculopathy, which includes micro- and macrovascular diabetic complications. Nicotinamide adenine dinucleotide phosphate oxidase (NOX) is a membrane-bound enzyme responsible for the development of reactive oxygen species in hyperglycaemia. Phosphorylation of the cytosolic components of NOX, such as p47phox, p67phox, and RAC-1, in hyperglycaemia is one of the important causes of conversion of oxygen to reactive oxygen. Overexpression of NOX in pathological conditions is associated with activation of aldose reductase, advanced glycation end products, protein kinase C and the hexosamine pathway. In addition, NOX also promotes the activation of inflammatory cytokines, such as TGF-β, TNF-α, NF-kβ, IL-6, and IL-18, the activation of endothelial growth factors, such as VEGF and FGF, hyperlipidaemia, and the deposition of collagen. Thus, overexpression of NOX is linked to the development of diabetic complications. The present review focuses on the role of NOX, its associated pathways, and various NOX inhibitors in the management and treatment of diabetic complications, such as diabetic nephropathy, retinopathy, neuropathy and cardiomyopathy.
Collapse
Affiliation(s)
- Ankit P Laddha
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L Mehta Road, Vile Parle (W), Mumbai, 400 056, India
| | - Yogesh A Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS, V. L Mehta Road, Vile Parle (W), Mumbai, 400 056, India.
| |
Collapse
|
16
|
Miller YI, Navia-Pelaez JM, Corr M, Yaksh TL. Lipid rafts in glial cells: role in neuroinflammation and pain processing. J Lipid Res 2020; 61:655-666. [PMID: 31862695 PMCID: PMC7193960 DOI: 10.1194/jlr.tr119000468] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/06/2019] [Indexed: 12/27/2022] Open
Abstract
Activation of microglia and astrocytes secondary to inflammatory processes contributes to the development and perpetuation of pain with a neuropathic phenotype. This pain state presents as a chronic debilitating condition and affects a large population of patients with conditions like rheumatoid arthritis and diabetes, or after surgery, trauma, or chemotherapy. Here, we review the regulation of lipid rafts in glial cells and the role they play as a key component of neuroinflammatory sensitization of central pain signaling pathways. In this context, we introduce the concept of an inflammaraft (i-raft), enlarged lipid rafts harboring activated receptors and adaptor molecules and serving as an organizing platform to initiate inflammatory signaling and the cellular response. Characteristics of the inflammaraft include increased relative abundance of lipid rafts in inflammatory cells, increased content of cholesterol per raft, and increased levels of inflammatory receptors, such as toll-like receptor (TLR)4, adaptor molecules, ion channels, and enzymes in lipid rafts. This inflammaraft motif serves an important role in the membrane assembly of protein complexes, for example, TLR4 dimerization. Operating within this framework, we demonstrate the involvement of inflammatory receptors, redox molecules, and ion channels in the inflammaraft formation and the regulation of cholesterol and sphingolipid metabolism in the inflammaraft maintenance and disruption. Strategies for targeting inflammarafts, without affecting the integrity of lipid rafts in noninflammatory cells, may lead to developing novel therapies for neuropathic pain states and other neuroinflammatory conditions.
Collapse
Affiliation(s)
- Yury I Miller
- Departments of MedicineUniversity of California San Diego, La Jolla, CA. mailto:
| | | | - Maripat Corr
- Departments of MedicineUniversity of California San Diego, La Jolla, CA
| | - Tony L Yaksh
- Anesthesiology,University of California San Diego, La Jolla, CA
| |
Collapse
|
17
|
NADPH oxidases: Pathophysiology and therapeutic potential in age-associated pulmonary fibrosis. Redox Biol 2020; 33:101541. [PMID: 32360174 PMCID: PMC7251244 DOI: 10.1016/j.redox.2020.101541] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/08/2020] [Accepted: 04/10/2020] [Indexed: 02/06/2023] Open
Abstract
Oxidative stress has been associated with a number of human fibrotic diseases, including idiopathic pulmonary fibrosis (IPF). Although oxidative stress is associated with both fibrosis and aging, the precise cellular sources(s) of reactive oxygen species (ROS) that contribute to the disease pathogenesis remain poorly understood. NADPH oxidase (Nox) enzymes are an evolutionarily conserved family, where their only known function is the production of ROS. A growing body of evidence supports a link between excessive Nox-derived ROS and numerous chronic diseases (including fibrotic disease), which is most prevalent among the elderly population. In this review, we examine the evidence for Nox isoforms in the pathogenesis of IPF, and the potential to target this enzyme family for the treatment of IPF and related fibrotic disorders. A better understanding of the Nox-mediated redox imbalance in aging may be critical to the development of more effective therapeutic strategies for age-associated fibrotic disorders. Strategies aimed at specifically blocking the source(s) of ROS through Nox inhibition may prove to be more effective as anti-fibrotic therapies, as compared to antioxidant approaches. This review also discusses the potential of Nox-targeting therapeutics currently in development.
Collapse
|
18
|
Guo D, Xu S, Yasen W, Zhang C, Shen J, Huang Y, Chen D, Zhu X. Tirapazamine-embedded polyplatinum(iv) complex: a prodrug combo for hypoxia-activated synergistic chemotherapy. Biomater Sci 2020; 8:694-701. [DOI: 10.1039/c9bm01640f] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A polyprodrug complex containing oxygen depleting chemodrugs and hypoxia-activated antitumor agents can serve as a promising drug delivery system for synergistic chemotherapy.
Collapse
Affiliation(s)
- Dongbo Guo
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Shuting Xu
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Wumaier Yasen
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Chuan Zhang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Jian Shen
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials
- Jiangsu Key Laboratory of Biomedical Materials
- College of Chemistry and Materials Science
- Nanjing Normal University
- Nanjing 210046
| | - Yu Huang
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Dong Chen
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| | - Xinyuan Zhu
- School of Chemistry and Chemical Engineering
- State Key Laboratory of Metal Matrix Composites
- Shanghai Jiao Tong University
- Shanghai 200240
- China
| |
Collapse
|
19
|
Ishii T, Warabi E. Mechanism of Rapid Nuclear Factor-E2-Related Factor 2 (Nrf2) Activation via Membrane-Associated Estrogen Receptors: Roles of NADPH Oxidase 1, Neutral Sphingomyelinase 2 and Epidermal Growth Factor Receptor (EGFR). Antioxidants (Basel) 2019; 8:antiox8030069. [PMID: 30889865 PMCID: PMC6466580 DOI: 10.3390/antiox8030069] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/07/2019] [Accepted: 03/13/2019] [Indexed: 12/12/2022] Open
Abstract
Membrane-associated estrogen receptors (ER)-α36 and G protein-coupled estrogen receptor (GPER) play important roles in the estrogen’s rapid non-genomic actions including stimulation of cell proliferation. Estrogen via these receptors induces rapid activation of transcription factor nuclear factor-E2-related factor 2 (Nrf2), a master regulator of detoxification and antioxidant systems, playing a key role in the metabolic reprogramming to support cell proliferation. This review highlights the possible mechanism underlying rapid Nrf2 activation via membrane-associated estrogen receptors by estrogen and phytoestrogens. Stimulation of ER-α36-GPER signaling complex rapidly induces Src-mediated transactivation of epidermal growth factor receptor (EGFR) leading to a kinase-mediated signaling cascade. We propose a novel hypothesis that ER-α36-GPER signaling initially induces rapid and temporal activation of NADPH oxidase 1 to generate superoxide, which subsequently activates redox-sensitive neutral sphingomyelinase 2 generating the lipid signaling mediator ceramide. Generation of ceramide is required for Ras activation and ceramide-protein kinase C ζ-casein kinase 2 (CK2) signaling. Notably, CK2 enhances chaperone activity of the Cdc37-Hsp90 complex supporting activation of various signaling kinases including Src, Raf and Akt (protein kinase B). Activation of Nrf2 may be induced by cooperation of two signaling pathways, (i) Nrf2 stabilization by direct phosphorylation by CK2 and (ii) EGFR-Ras-PI 3 kinase (PI3K)-Akt axis which inhibits glycogen synthase kinase 3β leading to enhanced nuclear transport and stability of Nrf2.
Collapse
Affiliation(s)
- Tetsuro Ishii
- Faculty of Medicine, University of Tsukuba, Tsukuba Ibaraki 305-8575, Japan.
| | - Eiji Warabi
- Faculty of Medicine, University of Tsukuba, Tsukuba Ibaraki 305-8575, Japan.
| |
Collapse
|
20
|
Parascandolo A, Laukkanen MO. Carcinogenesis and Reactive Oxygen Species Signaling: Interaction of the NADPH Oxidase NOX1-5 and Superoxide Dismutase 1-3 Signal Transduction Pathways. Antioxid Redox Signal 2019; 30:443-486. [PMID: 29478325 PMCID: PMC6393772 DOI: 10.1089/ars.2017.7268] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Revised: 02/21/2018] [Accepted: 02/22/2018] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE Reduction/oxidation (redox) balance could be defined as an even distribution of reduction and oxidation complementary processes and their reaction end products. There is a consensus that aberrant levels of reactive oxygen species (ROS), commonly observed in cancer, stimulate primary cell immortalization and progression of carcinogenesis. However, the mechanism how different ROS regulate redox balance is not completely understood. Recent Advances: In the current review, we have summarized the main signaling cascades inducing NADPH oxidase NOX1-5 and superoxide dismutase (SOD) 1-3 expression and their connection to cell proliferation, immortalization, transformation, and CD34+ cell differentiation in thyroid, colon, lung, breast, and hematological cancers. CRITICAL ISSUES Interestingly, many of the signaling pathways activating redox enzymes or mediating the effect of ROS are common, such as pathways initiated from G protein-coupled receptors and tyrosine kinase receptors involving protein kinase A, phospholipase C, calcium, and small GTPase signaling molecules. FUTURE DIRECTIONS The clarification of interaction of signal transduction pathways could explain how cells regulate redox balance and may even provide means to inhibit the accumulation of harmful levels of ROS in human pathologies.
Collapse
|
21
|
Sumimoto H, Minakami R, Miyano K. Soluble Regulatory Proteins for Activation of NOX Family NADPH Oxidases. Methods Mol Biol 2019; 1982:121-137. [PMID: 31172470 DOI: 10.1007/978-1-4939-9424-3_8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
NOX family NADPH oxidases deliberately produce reactive oxygen species and thus contribute to a variety of biological functions. Of seven members in the human family, the three oxidases NOX2, NOX1, and NOX3 form a heterodimer with p22phox and are regulated by soluble regulatory proteins: p47phox, its related organizer NOXO1; p67phox, its related activator NOXA1; p40phox; and the small GTPase Rac. Activation of the phagocyte oxidase NOX2 requires p47phox, p67phox, and GTP-bound Rac. In addition to these regulators, p40phox plays a crucial role when NOX2 is activated during phagocytosis. On the other hand, NOX1 activation prefers NOXO1 and NOXA1, although Rac is also involved. NOX3 constitutively produces superoxide, which is enhanced by regulatory proteins such as p47phox, NOXO1, and p67phox. Here we describe mechanisms for NOX activation with special attention to the soluble regulatory proteins.
Collapse
Affiliation(s)
- Hideki Sumimoto
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.
| | - Reiko Minakami
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kei Miyano
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
22
|
Nordzieke DE, Medraño-Fernandez I. The Plasma Membrane: A Platform for Intra- and Intercellular Redox Signaling. Antioxidants (Basel) 2018; 7:antiox7110168. [PMID: 30463362 PMCID: PMC6262572 DOI: 10.3390/antiox7110168] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 11/15/2018] [Accepted: 11/17/2018] [Indexed: 02/06/2023] Open
Abstract
Membranes are of outmost importance to allow for specific signal transduction due to their ability to localize, amplify, and direct signals. However, due to the double-edged nature of reactive oxygen species (ROS)—toxic at high concentrations but essential signal molecules—subcellular localization of ROS-producing systems to the plasma membrane has been traditionally regarded as a protective strategy to defend cells from unwanted side-effects. Nevertheless, specialized regions, such as lipid rafts and caveolae, house and regulate the activated/inhibited states of important ROS-producing systems and concentrate redox targets, demonstrating that plasma membrane functions may go beyond acting as a securing lipid barrier. This is nicely evinced by nicotinamide adenine dinucleotide phosphate (NADPH)-oxidases (NOX), enzymes whose primary function is to generate ROS and which have been shown to reside in specific lipid compartments. In addition, membrane-inserted bidirectional H2O2-transporters modulate their conductance precisely during the passage of the molecules through the lipid bilayer, ensuring time-scaled delivery of the signal. This review aims to summarize current evidence supporting the role of the plasma membrane as an organizing center that serves as a platform for redox signal transmission, particularly NOX-driven, providing specificity at the same time that limits undesirable oxidative damage in case of malfunction. As an example of malfunction, we explore several pathological situations in which an inflammatory component is present, such as inflammatory bowel disease and neurodegenerative disorders, to illustrate how dysregulation of plasma-membrane-localized redox signaling impacts normal cell physiology.
Collapse
Affiliation(s)
- Daniela E Nordzieke
- Institute of Microbiology and Genetics, Department of Genetics of Eukaryotic Microorganisms, Georg August University Göttingen, Grisebachstr. 8, D-37077 Göttingen, Germany.
| | - Iria Medraño-Fernandez
- Protein Transport and Secretion Unit, Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132 Milan, Italy.
| |
Collapse
|
23
|
Rezende F, Moll F, Walter M, Helfinger V, Hahner F, Janetzko P, Ringel C, Weigert A, Fleming I, Weissmann N, Kuenne C, Looso M, Rieger MA, Nawroth P, Fleming T, Brandes RP, Schröder K. The NADPH organizers NoxO1 and p47phox are both mediators of diabetes-induced vascular dysfunction in mice. Redox Biol 2018; 15:12-21. [PMID: 29195137 PMCID: PMC5723277 DOI: 10.1016/j.redox.2017.11.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 11/01/2017] [Accepted: 11/16/2017] [Indexed: 12/12/2022] Open
Abstract
AIM NADPH oxidases are important sources of reactive oxygen species (ROS). Several Nox homologues are present together in the vascular system but whether they exhibit crosstalk at the activity level is unknown. To address this, vessel function of knockout mice for the cytosolic Nox organizer proteins p47phox, NoxO1 and a p47phox-NoxO1-double knockout were studied under normal condition and during streptozotocin-induced diabetes. RESULTS In the mouse aorta, mRNA expression for NoxO1 was predominant in smooth muscle and endothelial cells, whereas p47phox was markedly expressed in adventitial cells comprising leukocytes and tissue resident macrophages. Knockout of either NoxO1 or p47phox resulted in lower basal blood pressure. Deletion of any of the two subunits also prevented diabetes-induced vascular dysfunction. mRNA expression analysis by MACE (Massive Analysis of cDNA ends) identified substantial gene expression differences between the mouse lines and in response to diabetes. Deletion of p47phox induced inflammatory activation with increased markers of myeloid cells and cytokine and chemokine induction. In contrast, deletion of NoxO1 resulted in an attenuated interferon gamma signature and reduced expression of genes related to antigen presentation. This aspect was also reflected by a reduced number of circulating lymphocytes in NoxO1-/- mice. INNOVATION AND CONCLUSION ROS production stimulated by NoxO1 and p47phox limit endothelium-dependent relaxation and maintain blood pressure in mice. However, NoxO1 and p47phox cannot substitute each other despite their similar effect on vascular function. Deletion of NoxO1 induced an anti-inflammatory phenotype, whereas p47phox deletion rather elicited a hyper-inflammatory response.
Collapse
Affiliation(s)
- Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt, Germany
| | - Franziska Moll
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt, Germany
| | - Maria Walter
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany
| | - Valeska Helfinger
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany
| | - Fabian Hahner
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany
| | - Patrick Janetzko
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany
| | - Christian Ringel
- Institute for Patho Biochemistry, Goethe University, Frankfurt, Germany
| | - Andreas Weigert
- Institute for Patho Biochemistry, Goethe University, Frankfurt, Germany
| | - Ingrid Fleming
- Institute for Vascular Signaling, Goethe-University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt, Germany
| | - Norbert Weissmann
- University of Giessen and Marburg Lung Center, German Center for Lung Research (DZL), Giessen, Germany
| | - Carsten Kuenne
- Max-Planck-Institute for Heart and Lung Research, Bioinformatics Core Facility, Bad Nauheim, Germany
| | - Mario Looso
- Max-Planck-Institute for Heart and Lung Research, Bioinformatics Core Facility, Bad Nauheim, Germany; German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt, Germany
| | - Michael A Rieger
- Department of Medicine, Hematology/Oncology, Goethe-University, Frankfurt, Germany
| | - Peter Nawroth
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Joint Division Molecular Metabolic Control, German Cancer Research Center (DKFZ) Heidelberg Center for Molecular Biology (ZMBH) and University Hospital Heidelberg University, Heidelberg, Germany; Institute for Diabetes and Cancer IDC Helmholtz Center Munich and Joint Heidelberg-IDC Translational Diabetes Program, Neuherberg, Germany
| | - Thomas Fleming
- Department of Internal Medicine I and Clinical Chemistry, University Hospital Heidelberg, Heidelberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany; Joint Division Molecular Metabolic Control, German Cancer Research Center (DKFZ) Heidelberg Center for Molecular Biology (ZMBH) and University Hospital Heidelberg University, Heidelberg, Germany; Institute for Diabetes and Cancer IDC Helmholtz Center Munich and Joint Heidelberg-IDC Translational Diabetes Program, Neuherberg, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt, Germany.
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt, Germany; German Center of Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt, Germany.
| |
Collapse
|
24
|
Gao L, Hu Y, Li J. Pigment epithelium-derived factor protects human glomerular mesangial cells from diabetes via NOXO1-iNOS suppression. Mol Med Rep 2017; 16:7855-7863. [DOI: 10.3892/mmr.2017.7563] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 09/05/2017] [Indexed: 11/05/2022] Open
|
25
|
Yu W, Ji W, Mi L, Lin C. Mechanisms of N‑acetylcysteine in reducing monocrotaline‑induced pulmonary hypertension in rats: Inhibiting the expression of Nox1 in pulmonary vascular smooth muscle cells. Mol Med Rep 2017; 16:6148-6155. [PMID: 28849167 DOI: 10.3892/mmr.2017.7326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 05/30/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate the impact of N‑acetylcysteine (NAC) on the expression of reduced nicotinamide adenine dinucleotide phosphate oxidase 1 (Nox1), and the proliferation and apoptosis of pulmonary artery smooth muscle cells (PASMCs) in rats exhibiting monocrotaline (MCT)‑induced pulmonary hypertension, and to investigate the possible mechanisms and treatment roles of NAC in pulmonary vascular remodeling (PVR). A total of 18 Wistar rats were randomly divided into three groups: The control (C) group; the MCT (M) group; and the NAC (N) group. The right ventricular hypertrophy index (RVHI) and other indicators were recorded 6 weeks subsequently. Groups C and M were divided into two subgroups: Groups C1 and M1 (control); and group C2 and M2 group (treated with ML171). Group N was not sub‑divided. PASMCs were isolated, and the vascular remodeling and Nox1 positioning were observed. The expression of Nox mRNA in each group, and the proliferation, apoptosis, and superoxide dismutase (SOD) activity of PASMCs, prior to and following the ML171 treatment, were measured. NAC was able to decrease RVHI and other indicators (P<0.001). The mRNA expression of Nox1 and Nox4 in group M was significantly increased compared with group C (P<0.05), and NAC was able to significantly decrease the expression of these two factors in lung tissue (P<0.001). MCT‑PASMCs exhibited differences in Nox1 mRNA expression (P<0.001), and the total SOD activity was Nox1‑dependently increased (r=0.949; P<0.001). NAC was able to decrease Nox1‑derived reactive oxygen species in PASMCs, thereby improving PVR. Nox1 was able to increase SOD activity, thereby demonstrating its positive effect on the proliferation of MCT‑PASMCs.
Collapse
Affiliation(s)
- Wencheng Yu
- Department of Respiratory Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Weina Ji
- Department of Respiratory Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Liyun Mi
- Department of Respiratory Diseases, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 266003, P.R. China
| | - Chen Lin
- Department of Respiratory Diseases, Shenli Oilfield Central Hospital, Dongying, Liaoning 257064, P.R. China
| |
Collapse
|
26
|
Schröder K, Weissmann N, Brandes RP. Organizers and activators: Cytosolic Nox proteins impacting on vascular function. Free Radic Biol Med 2017; 109:22-32. [PMID: 28336130 DOI: 10.1016/j.freeradbiomed.2017.03.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/24/2017] [Accepted: 03/14/2017] [Indexed: 01/25/2023]
Abstract
NADPH oxidases of the Nox family are important enzymatic sources of reactive oxygen species (ROS) in the cardiovascular system. Of the 7 members of the Nox family, at least three depend for their activation on specific cytosolic proteins. These are p47phox and its homologue NoxO1 and p67phox and its homologue NoxA1. Also the Rho-GTPase Rac is important but as this protein has many additional functions, it will not be covered here. The Nox1 enzyme is preferentially activated by the combination of NoxO1 with NoxA1, whereas Nox2 gains highest activity with p47phox together with p67phox. As p47phox, different to NoxO1 contains an auto inhibitory region it has to be phosphorylated prior to complex formation. In the cardio-vascular system, all cytosolic Nox proteins are expressed but the evidence for their contribution to ROS production is not well established. Most data have been collected for p47phox, whereas NoxA1 has basically not yet been studied. In this article the specific aspects of cytosolic Nox proteins in the cardiovascular system with respect to Nox activation, their expression and their importance will be reviewed. Finally, it will be discussed whether cytosolic Nox proteins are suitable pharmacological targets to tamper with vascular ROS production.
Collapse
Affiliation(s)
- Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany.
| | - Norbert Weissmann
- Excellence Cluster Cardiopulmonary System, University of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Justus-Liebig-University, 35392 Giessen, Germany
| | - Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany
| |
Collapse
|
27
|
de Souza Santos M, Salomon D, Orth K. T3SS effector VopL inhibits the host ROS response, promoting the intracellular survival of Vibrio parahaemolyticus. PLoS Pathog 2017. [PMID: 28640881 PMCID: PMC5481031 DOI: 10.1371/journal.ppat.1006438] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The production of antimicrobial reactive oxygen species by the nicotinamide dinucleotide phosphate (NADPH) oxidase complex is an important mechanism for control of invading pathogens. Herein, we show that the gastrointestinal pathogen Vibrio parahaemolyticus counteracts reactive oxygen species (ROS) production using the Type III Secretion System 2 (T3SS2) effector VopL. In the absence of VopL, intracellular V. parahaemolyticus undergoes ROS-dependent filamentation, with concurrent limited growth. During infection, VopL assembles actin into non-functional filaments resulting in a dysfunctional actin cytoskeleton that can no longer mediate the assembly of the NADPH oxidase at the cell membrane, thereby limiting ROS production. This is the first example of how a T3SS2 effector contributes to the intracellular survival of V. parahaemolyticus, supporting the establishment of a protective intracellular replicative niche.
Collapse
Affiliation(s)
- Marcela de Souza Santos
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Dor Salomon
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - Kim Orth
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
- * E-mail:
| |
Collapse
|
28
|
C-terminal tail of NADPH oxidase organizer 1 (Noxo1) mediates interaction with NADPH oxidase activator (Noxa1) in the NOX1 complex. Biochem Biophys Res Commun 2017. [PMID: 28625920 DOI: 10.1016/j.bbrc.2017.06.083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
NOX1 (NADPH oxidase) similar to phagocyte NADPH oxidase, is expressed mainly in the colon epithelium and it is responsible for host defense against microbial infections by generating ROS (reactive oxygen species). NOX1 is activated by two regulatory cytosolic proteins that form a hetero-dimer, Noxo1 (NOX organizer 1) and Noxa1 (NOX activator 1). The interaction between Noxa1 and Noxo1 is critical for activating NOX1. However no structural studies for interaction between Noxa1 and Noxo1 has not been reported till date. Here, we studied the inter-molecular interaction between the SH3 domain of Noxa1 and Noxo1 using pull-down assay and NMR spectroscopy. 15N/13C-labeled SH3 domain of Noxa1 has been purified for hetero-nuclear NMR experiments (HNCACB, CBCACONH, HNCA, HNCO, and HSQC). TALOS analysis using backbone assignment data of the Noxa1 SH3 domain showed that the structure primarily consists of β-sheets. Data from pull-down assay between the Noxo1 and Noxa1 showed that the SH3 domains (Noxa1) is responsible for interaction with Noxo1 C-terminal tail harboring proline rich region (PRR). The concentration-dependent titration of the Noxo1 C-terminal tail to Noxa1 shows that Noxo1 particularly in the RT loop: Q407*, H408, S409, A412*, G414*, E416, D417, L418, and F420; n-Src loop: C430, E431*, V432*, A435, W436, and L437; and terminal region: I447; F448*, F452* and V454 interact with Noxa1. Our results will provide a detailed understanding for interaction between Noxa1 and Noxo1 at the molecular level, providing insights into their cytoplasmic activity-mediated functioning as well as regulatory role of C-terminal tail of Noxo1 in the NOX1 complex.
Collapse
|
29
|
Jones RM, Neish AS. Redox signaling mediated by the gut microbiota. Free Radic Biol Med 2017; 105:41-47. [PMID: 27989756 DOI: 10.1016/j.freeradbiomed.2016.10.495] [Citation(s) in RCA: 125] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/17/2016] [Accepted: 10/21/2016] [Indexed: 12/27/2022]
Abstract
The microbiota that inhabits the mammalian intestine can influence a range of physiological functions, including the modulation of immune responses, enhancement epithelial barrier function, and the stimulation of cell proliferation. While the mechanisms by which commensal prokaryotes stimulate immune signaling networks are well-characterized, less is known about the mechanistic control over homeostatic pathways within tissues. Recent reports by our research group have demonstrated that contact between the gut epithelia and some groups of enteric commensal bacteria prompts the rapid generation of reactive oxygen species (ROS) within host cells. Whereas the bacterial-induced production of ROS in phagocytes in response to ligand binding to Formyl Peptide Receptors (FPRs) and ensuing activation of NADPH oxidase 2 (Nox2) is a well-defined mechanism, ROS generated by other cell types such as intestinal epithelia in response to microbial signals via FPRs and the NADPH oxidase 1 (Nox1) is less appreciated. Importantly, enzymatically generated ROS have been shown to function as second messengers in many signal transduction pathways via the transient oxidative activity on sensor proteins bearing oxidant-sensitive thiol groups. Examples of redox sensitive proteins include tyrosine phosphatases that serve as regulators of MAPK pathways, focal adhesion kinase, as well as components involved NF-kB activation. Here, we review the leading edge discoveries gleaned from investigations that focus on microbial-induced generation of ROS and their functional effects on host physiology. These studies identify the functional molecular elements and mechanistic events that mediate the established effects of the normal microbiota on intestinal physiology.
Collapse
Affiliation(s)
- Rheinallt M Jones
- Department of Pediatrics, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michaels St, Room 105-L, Atlanta, GA 30322, United States
| | - Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michaels St, Room 105-L, Atlanta, GA 30322, United States.
| |
Collapse
|
30
|
Juhasz A, Markel S, Gaur S, Liu H, Lu J, Jiang G, Wu X, Antony S, Wu Y, Melillo G, Meitzler JL, Haines DC, Butcher D, Roy K, Doroshow JH. NADPH oxidase 1 supports proliferation of colon cancer cells by modulating reactive oxygen species-dependent signal transduction. J Biol Chem 2017; 292:7866-7887. [PMID: 28330872 PMCID: PMC5427267 DOI: 10.1074/jbc.m116.768283] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) play a critical role in cell signaling and proliferation. NADPH oxidase 1 (NOX1), a membrane-bound flavin dehydrogenase that generates O2˙̄, is highly expressed in colon cancer. To investigate the role that NOX1 plays in colon cancer growth, we used shRNA to decrease NOX1 expression stably in HT-29 human colon cancer cells. The 80–90% decrease in NOX1 expression achieved by RNAi produced a significant decline in ROS production and a G1/S block that translated into a 2–3-fold increase in tumor cell doubling time without increased apoptosis. The block at the G1/S checkpoint was associated with a significant decrease in cyclin D1 expression and profound inhibition of mitogen-activated protein kinase (MAPK) signaling. Decreased steady-state MAPK phosphorylation occurred concomitant with a significant increase in protein phosphatase activity for two colon cancer cell lines in which NOX1 expression was knocked down by RNAi. Diminished NOX1 expression also contributed to decreased growth, blood vessel density, and VEGF and hypoxia-inducible factor 1α (HIF-1α) expression in HT-29 xenografts initiated from NOX1 knockdown cells. Microarray analysis, supplemented by real-time PCR and Western blotting, revealed that the expression of critical regulators of cell proliferation and angiogenesis, including c-MYC, c-MYB, and VEGF, were down-regulated in association with a decline in hypoxic HIF-1α protein expression downstream of silenced NOX1 in both colon cancer cell lines and xenografts. These studies suggest a role for NOX1 in maintaining the proliferative phenotype of some colon cancers and the potential of NOX1 as a therapeutic target in this disease.
Collapse
Affiliation(s)
- Agnes Juhasz
- From the Developmental Therapeutics Branch of the Center for Cancer Research
| | - Susan Markel
- the Department of Medical Oncology and Therapeutics Research and
| | - Shikha Gaur
- the Department of Medical Oncology and Therapeutics Research and
| | - Han Liu
- the Division of Cancer Treatment and Diagnosis, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - Jiamo Lu
- From the Developmental Therapeutics Branch of the Center for Cancer Research
| | - Guojian Jiang
- From the Developmental Therapeutics Branch of the Center for Cancer Research
| | - Xiwei Wu
- the Bioinformatics Group, City of Hope Comprehensive Cancer Center, Duarte, California 91010
| | - Smitha Antony
- From the Developmental Therapeutics Branch of the Center for Cancer Research
| | - Yongzhong Wu
- From the Developmental Therapeutics Branch of the Center for Cancer Research
| | - Giovanni Melillo
- the Developmental Therapeutics Program, SAIC-Frederick, Inc., NCI at Frederick, Frederick, Maryland 21702, and
| | - Jennifer L Meitzler
- From the Developmental Therapeutics Branch of the Center for Cancer Research
| | - Diana C Haines
- the Pathology/Histotechnology Laboratory, Leidos, Inc./Frederick National Laboratory for Cancer Research, NCI, Frederick, Maryland 21702
| | - Donna Butcher
- the Pathology/Histotechnology Laboratory, Leidos, Inc./Frederick National Laboratory for Cancer Research, NCI, Frederick, Maryland 21702
| | - Krishnendu Roy
- the Division of Cancer Treatment and Diagnosis, NCI, National Institutes of Health, Bethesda, Maryland 20892
| | - James H Doroshow
- From the Developmental Therapeutics Branch of the Center for Cancer Research, .,the Division of Cancer Treatment and Diagnosis, NCI, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
31
|
Ma MW, Wang J, Zhang Q, Wang R, Dhandapani KM, Vadlamudi RK, Brann DW. NADPH oxidase in brain injury and neurodegenerative disorders. Mol Neurodegener 2017; 12:7. [PMID: 28095923 PMCID: PMC5240251 DOI: 10.1186/s13024-017-0150-7] [Citation(s) in RCA: 299] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2016] [Accepted: 01/05/2017] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress is a common denominator in the pathology of neurodegenerative disorders such as Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, amyotrophic lateral sclerosis, and multiple sclerosis, as well as in ischemic and traumatic brain injury. The brain is highly vulnerable to oxidative damage due to its high metabolic demand. However, therapies attempting to scavenge free radicals have shown little success. By shifting the focus to inhibit the generation of damaging free radicals, recent studies have identified NADPH oxidase as a major contributor to disease pathology. NADPH oxidase has the primary function to generate free radicals. In particular, there is growing evidence that the isoforms NOX1, NOX2, and NOX4 can be upregulated by a variety of neurodegenerative factors. The majority of recent studies have shown that genetic and pharmacological inhibition of NADPH oxidase enzymes are neuroprotective and able to reduce detrimental aspects of pathology following ischemic and traumatic brain injury, as well as in chronic neurodegenerative disorders. This review aims to summarize evidence supporting the role of NADPH oxidase in the pathology of these neurological disorders, explores pharmacological strategies of targeting this major oxidative stress pathway, and outlines obstacles that need to be overcome for successful translation of these therapies to the clinic.
Collapse
Affiliation(s)
- Merry W Ma
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Jing Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ruimin Wang
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Krishnan M Dhandapani
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA.,Department of Neurosurgery, Medical College of Georgia, Augusta University, 1120 Fifteenth Street, Augusta, GA, 30912, USA
| | - Ratna K Vadlamudi
- Department of Obstetrics and Gynecology, University of Texas Health Science Center, 7703 Medical Drive, San Antonio, TX, 78229, USA
| | - Darrell W Brann
- Charlie Norwood VA Medical Center, One Freedom Way, Augusta, GA, 30904, USA. .,Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, 1120 Fifteenth Street, Augusta, GA, 30912, USA.
| |
Collapse
|
32
|
Shanmugasundaram K, Block K. Renal Carcinogenesis, Tumor Heterogeneity, and Reactive Oxygen Species: Tactics Evolved. Antioxid Redox Signal 2016; 25:685-701. [PMID: 27287984 PMCID: PMC5069729 DOI: 10.1089/ars.2015.6569] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 06/07/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE The number of kidney cancers is growing 3-5% each year due to unknown etiologies. Intra- and inter-tumor mediators increase oxidative stress and drive tumor heterogeneity. Recent Advances: Technology advancement in state-of-the-art instrumentation and methodologies allows researchers to detect and characterize global landscaping modifications in genes, proteins, and pathophysiology patterns at the single-cell level. CRITICAL ISSUES We postulate that the sources of reactive oxygen species (ROS) and their activation within subcellular compartments will change over a timeline of tumor evolvement and contribute to tumor heterogeneity. Therefore, the complexity of intracellular changes within a tumor and ROS-induced tumor heterogeneity coupled to the advancement of detecting these events globally are limited at the level of data collection, organization, and interpretation using software algorithms and bioinformatics. FUTURE DIRECTIONS Integrative and collaborative research, combining the power of numbers with careful experimental design, protocol development, and data interpretation, will translate cancer biology and therapeutics to a heightened level or leave the abundant raw data as stagnant and underutilized. Antioxid. Redox Signal. 25, 685-701.
Collapse
Affiliation(s)
| | - Karen Block
- Department of Medicine, University of Texas Health Science Center, San Antonio, Texas
- South Texas Veterans Health Care System, Audie L. Murphy Memorial Hospital Division, San Antonio, Texas
| |
Collapse
|
33
|
Brandes RP, Harenkamp S, Schürmann C, Josipovic I, Rashid B, Rezende F, Löwe O, Moll F, Epah J, Eresch J, Nayak A, Kopaliani I, Penski C, Mittelbronn M, Weissmann N, Schröder K. The Cytosolic NADPH Oxidase Subunit NoxO1 Promotes an Endothelial Stalk Cell Phenotype. Arterioscler Thromb Vasc Biol 2016; 36:1558-65. [PMID: 27283741 DOI: 10.1161/atvbaha.116.307132] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 05/31/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Reactive oxygen species generated by nicotinamide adenine dinucleotide phosphate (NADPH) oxidases contribute to angiogenesis and vascular repair. NADPH oxidase organizer 1 (NoxO1) is a cytosolic protein facilitating assembly of constitutively active NADPH oxidases. We speculate that NoxO1 also contributes to basal reactive oxygen species formation in the vascular system and thus modulates angiogenesis. APPROACH AND RESULTS A NoxO1 knockout mouse was generated, and angiogenesis was studied in cultured cells and in vivo. Angiogenesis of the developing retina and after femoral artery ligation was increased in NoxO1(-/-) when compared with wild-type animals. Spheroid outgrowth assays revealed greater angiogenic capacity of NoxO1(-/-) lung endothelial cells (LECs) and a more tip-cell-like phenotype than wild-type LECs. Usually signaling by the Notch pathway switches endothelial cells from a tip into a stalk cell phenotype. NoxO1(-/-) LECs exhibited attenuated Notch signaling as a consequence of an attenuated release of the Notch intracellular domain on ligand stimulation. This release is mediated by proteolytic cleavage involving the α-secretase ADAM17. For maximal activity, ADAM17 has to be oxidized, and overexpression of NoxO1 promoted this mode of activation. Moreover, the activity of ADAM17 was reduced in NoxO1(-/-) LECs when compared with wild-type LECs. CONCLUSIONS NoxO1 stimulates α-secretase activity probably through reactive oxygen species-mediated oxidation. Deletion of NoxO1 attenuates Notch signaling and thereby promotes a tip-cell phenotype that results in increased angiogenesis.
Collapse
Affiliation(s)
- Ralf P Brandes
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Sabine Harenkamp
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Christoph Schürmann
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Ivana Josipovic
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Beliza Rashid
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Flavia Rezende
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Oliver Löwe
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Franziska Moll
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Jeremy Epah
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Jeanette Eresch
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Arnab Nayak
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Irakli Kopaliani
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Cornelia Penski
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Michel Mittelbronn
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Norbert Weissmann
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.)
| | - Katrin Schröder
- From the Institute for Cardiovascular Physiology (R.P.B., S.H., C.S., I.J., B.R., F.R., O.L., F.M., J.E., K.S.), Pharmazentrum Frankfurt (J.E.), Institute for Biochemistry II (A.N.), and Neurological Institute (Edinger Institute) (C.P., M.M.), Goethe University, Frankfurt, Germany; Department of Physiology, Medical Faculty, TU Dresden, Dresden, Germany (I.K.); Justus-Liebig Universität Giessen, Giessen, Germany (N.W.); and German Center for Cardiovascular Research (DZHK), Partner Site RheinMain, Frankfurt, Germany (R.P.B., C.S., I.J., F.R., O.L., F.M., K.S.).
| |
Collapse
|
34
|
Liang S, Kisseleva T, Brenner DA. The Role of NADPH Oxidases (NOXs) in Liver Fibrosis and the Activation of Myofibroblasts. Front Physiol 2016; 7:17. [PMID: 26869935 PMCID: PMC4735448 DOI: 10.3389/fphys.2016.00017] [Citation(s) in RCA: 145] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 01/12/2016] [Indexed: 12/18/2022] Open
Abstract
Chronic liver injury, resulted from different etiologies (e.g., virus infection, alcohol abuse, nonalcoholic steatohepatitis (NASH) and cholestasis) can lead to liver fibrosis characterized by the excess accumulation of extracellular matrix (ECM) proteins (e.g., type I collagen). Hepatic myofibroblasts that are activated upon liver injury are the key producers of ECM proteins, contributing to both the initiation and progression of liver fibrosis. Hepatic stellate cells (HSCs) and to a lesser extent, portal fibroblast, are believed to be the precursor cells that give rise to hepatic myofibroblasts in response to liver injury. Although, much progress has been made toward dissecting the lineage origin of myofibroblasts, how these cells are activated and become functional producers of ECM proteins remains incompletely understood. Activation of myofibroblasts is a complex process that involves the interactions between parenchymal and non-parenchymal cells, which drives the phenotypic change of HSCs from a quiescent stage to a myofibroblastic and active phenotype. Accumulating evidence has suggested a critical role of NADPH oxidase (NOX), a multi-component complex that catalyzes reactions from molecular oxygen to reactive oxygen species (ROS), in the activation process of hepatic myofibroblasts. NOX isoforms, including NOX1, NOX2 and NOX4, and NOX-derived ROS, have all been implicated to regulate HSC activation and hepatocyte apoptosis, both of which are essential steps for initiating liver fibrosis. This review highlights the importance of NOX isoforms in hepatic myofibroblast activation and the progression of liver fibrosis, and also discusses the therapeutic potential of targeting NOXs for liver fibrosis and associated hepatic diseases.
Collapse
Affiliation(s)
- Shuang Liang
- Department of Surgery, University of California, San DiegoLa Jolla, CA, USA; Department of Medicine, University of California, San DiegoLa Jolla, CA, USA
| | - Tatiana Kisseleva
- Department of Surgery, University of California, San Diego La Jolla, CA, USA
| | - David A Brenner
- Department of Medicine, University of California, San Diego La Jolla, CA, USA
| |
Collapse
|
35
|
Wieczfinska J, Sokolowska M, Pawliczak R. NOX Modifiers-Just a Step Away from Application in the Therapy of Airway Inflammation? Antioxid Redox Signal 2015; 23:428-45. [PMID: 24383678 PMCID: PMC4543397 DOI: 10.1089/ars.2013.5783] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
SIGNIFICANCE NADPH oxidase (NOX) enzymes, which are widely expressed in different airway cell types, not only contribute to the maintenance of physiological processes in the airways but also participate in the pathogenesis of many acute and chronic diseases. Therefore, the understanding of NOX isoform regulation, expression, and the manner of their potent inhibition might lead to effective therapeutic approaches. RECENT ADVANCES The study of the role of NADPH oxidases family in airway physiology and pathophysiology should be considered as a work in progress. While key questions still remain unresolved, there is significant progress in terms of our understanding of NOX importance in airway diseases as well as a more efficient way of using NOX modifiers in human settings. CRITICAL ISSUES Agents that modify the activity of NADPH enzyme components would be considered useful tools in the treatment of various airway diseases. Nevertheless, profound knowledge of airway pathology, as well as the mechanisms of NOX regulation is needed to develop potent but safe NOX modifiers. FUTURE DIRECTIONS Many compounds seem to be promising candidates for development into useful therapeutic agents, but their clinical potential is yet to be demonstrated. Further analysis of basic mechanisms in human settings, high-throughput compound scanning, clinical trials with new and existing molecules, and the development of new drug delivery approaches are the main directions of future studies on NOX modifiers. In this article, we discuss the current knowledge with regard to NOX isoform expression and regulation in airway inflammatory diseases as well as the aptitudes and therapeutic potential of NOX modifiers.
Collapse
Affiliation(s)
- Joanna Wieczfinska
- 1 Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz , Lodz, Poland
| | - Milena Sokolowska
- 2 Critical Care Medicine Department, Clinical Center, National Institutes of Health , Bethesda, Maryland
| | - Rafal Pawliczak
- 1 Department of Immunopathology, Faculty of Biomedical Sciences and Postgraduate Training, Medical University of Lodz , Lodz, Poland
| |
Collapse
|
36
|
Abstract
The mechanism by which reactive oxygen species (ROS) are produced by tumour cells remained incompletely understood until the discovery over the last 15 years of the family of NADPH oxidases (NOXs 1–5 and dual oxidases DUOX1/2) which are structural homologues of gp91phox, the major membrane-bound component of the respiratory burst oxidase of leucocytes. Knowledge of the roles of the NOX isoforms in cancer is rapidly expanding. Recent evidence suggests that both NOX1 and DUOX2 species produce ROS in the gastrointestinal tract as a result of chronic inflammatory stress; cytokine induction (by interferon-γ, tumour necrosis factor α, and interleukins IL-4 and IL-13) of NOX1 and DUOX2 may contribute to the development of colorectal and pancreatic carcinomas in patients with inflammatory bowel disease and chronic pancreatitis, respectively. NOX4 expression is increased in pre-malignant fibrotic states which may lead to carcinomas of the lung and liver. NOX5 is highly expressed in malignant melanomas, prostate cancer and Barrett's oesophagus-associated adenocarcinomas, and in the last it is related to chronic gastro-oesophageal reflux and inflammation. Over-expression of functional NOX proteins in many tissues helps to explain tissue injury and DNA damage from ROS that accompany pre-malignant conditions, as well as elucidating the potential mechanisms of NOX-related damage that contribute to both the initiation and the progression of a wide range of solid and haematopoietic malignancies.
Collapse
|
37
|
Vlahos R, Selemidis S. NADPH oxidases as novel pharmacologic targets against influenza A virus infection. Mol Pharmacol 2014; 86:747-59. [PMID: 25301784 DOI: 10.1124/mol.114.095216] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/14/2025] Open
Abstract
Influenza A viruses represent a major global health care challenge, with imminent pandemics, emerging antiviral resistance, and long lag times for vaccine development, raising a pressing need for novel pharmacologic strategies that ideally target the pathology irrespective of the infecting strain. Reactive oxygen species (ROS) pervade all facets of cell biology with both detrimental and protective properties. Indeed, there is compelling evidence that activation of the NADPH oxidase 2 (NOX2) isoform of the NADPH oxidase family of ROS-producing enzymes promotes lung oxidative stress, inflammation, injury, and dysfunction resulting from influenza A viruses of low to high pathogenicity, as well as impeding virus clearance. By contrast, the dual oxidase isoforms produce ROS that provide vital protective antiviral effects for the host. In this review, we propose that inhibitors of NOX2 are better alternatives than broad-spectrum antioxidant approaches for treatment of influenza pathologies, for which clinical efficacy may have been limited owing to poor bioavailability and inadvertent removal of beneficial ROS. Finally, we briefly describe the current suite of NADPH oxidase inhibitors and the molecular features of the NADPH oxidase enzymes that could be exploited by drug discovery for development of more specific and novel inhibitors to prevent or treat disease caused by influenza.
Collapse
Affiliation(s)
- Ross Vlahos
- Respiratory Research Group, Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne (R.V.), and Oxidant and Inflammation Biology Group, Department of Pharmacology, Monash University (S.S.), Victoria, Australia
| | - Stavros Selemidis
- Respiratory Research Group, Lung Health Research Centre, Department of Pharmacology and Therapeutics, University of Melbourne (R.V.), and Oxidant and Inflammation Biology Group, Department of Pharmacology, Monash University (S.S.), Victoria, Australia
| |
Collapse
|
38
|
Brandes RP, Weissmann N, Schröder K. Nox family NADPH oxidases: Molecular mechanisms of activation. Free Radic Biol Med 2014; 76:208-26. [PMID: 25157786 DOI: 10.1016/j.freeradbiomed.2014.07.046] [Citation(s) in RCA: 495] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 07/29/2014] [Accepted: 07/30/2014] [Indexed: 11/21/2022]
Abstract
NADPH oxidases of the Nox family are important enzymatic sources of reactive oxygen species (ROS). Numerous homologue-specific mechanisms control the activity of this enzyme family involving calcium, free fatty acids, protein-protein interactions, intracellular trafficking, and posttranslational modifications such as phosphorylation, acetylation, or sumoylation. After a brief review on the classic pathways of Nox activation, this article will focus on novel mechanisms of homologue-specific activity control and on cell-specific aspects which govern Nox activity. From these findings of the recent years it must be concluded that the activity control of Nox enzymes is much more complex than anticipated. Moreover, depending on the cellular activity state, Nox enzymes are selectively activated or inactivated. The complex upstream signaling aspects of these events make the development of "intelligent" Nox inhibitors plausible, which selectively attenuate disease-related Nox-mediated ROS formation without altering physiological signaling ROS. This approach might be of relevance for Nox-mediated tissue injury in ischemia-reperfusion and inflammation and also for chronic Nox overactivation as present in cancer initiation and cardiovascular disease.
Collapse
Affiliation(s)
- Ralf P Brandes
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany.
| | - Norbert Weissmann
- ECCPS, Justus-Liebig-Universität, Member of the DZL, Giessen, Germany
| | - Katrin Schröder
- Institut für Kardiovaskuläre Physiologie, Goethe-Universität Frankfurt, Frankfurt, Germany
| |
Collapse
|
39
|
NADPH oxidases: an overview from structure to innate immunity-associated pathologies. Cell Mol Immunol 2014; 12:5-23. [PMID: 25263488 DOI: 10.1038/cmi.2014.89] [Citation(s) in RCA: 661] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 12/11/2022] Open
Abstract
Oxygen-derived free radicals, collectively termed reactive oxygen species (ROS), play important roles in immunity, cell growth, and cell signaling. In excess, however, ROS are lethal to cells, and the overproduction of these molecules leads to a myriad of devastating diseases. The key producers of ROS in many cells are the NOX family of NADPH oxidases, of which there are seven members, with various tissue distributions and activation mechanisms. NADPH oxidase is a multisubunit enzyme comprising membrane and cytosolic components, which actively communicate during the host responses to a wide variety of stimuli, including viral and bacterial infections. This enzymatic complex has been implicated in many functions ranging from host defense to cellular signaling and the regulation of gene expression. NOX deficiency might lead to immunosuppression, while the intracellular accumulation of ROS results in the inhibition of viral propagation and apoptosis. However, excess ROS production causes cellular stress, leading to various lethal diseases, including autoimmune diseases and cancer. During the later stages of injury, NOX promotes tissue repair through the induction of angiogenesis and cell proliferation. Therefore, a complete understanding of the function of NOX is important to direct the role of this enzyme towards host defense and tissue repair or increase resistance to stress in a timely and disease-specific manner.
Collapse
|
40
|
Streeter J, Schickling BM, Jiang S, Stanic B, Thiel WH, Gakhar L, Houtman JCD, Miller FJ. Phosphorylation of Nox1 regulates association with NoxA1 activation domain. Circ Res 2014; 115:911-8. [PMID: 25228390 DOI: 10.1161/circresaha.115.304267] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Activation of Nox1 initiates redox-dependent signaling events crucial in the pathogenesis of vascular disease. Selective targeting of Nox1 is an attractive potential therapy, but requires a better understanding of the molecular modifications controlling its activation. OBJECTIVE To determine whether posttranslational modifications of Nox1 regulate its activity in vascular cells. METHODS AND RESULTS We first found evidence that Nox1 is phosphorylated in multiple models of vascular disease. Next, studies using mass spectroscopy and a pharmacological inhibitor demonstrated that protein kinase C-beta1 mediates phosphorylation of Nox1 in response to tumor necrosis factor-α. siRNA-mediated silencing of protein kinase C-beta1 abolished tumor necrosis factor-α-mediated reactive oxygen species production and vascular smooth muscle cell migration. Site-directed mutagenesis and isothermal titration calorimetry indicated that protein kinase C-beta1 phosphorylates Nox1 at threonine 429. Moreover, Nox1 threonine 429 phosphorylation facilitated the association of Nox1 with the NoxA1 activation domain and was necessary for NADPH oxidase complex assembly, reactive oxygen species production, and vascular smooth muscle cell migration. CONCLUSIONS We conclude that protein kinase C-beta1 phosphorylation of threonine 429 regulates activation of Nox1 NADPH oxidase.
Collapse
Affiliation(s)
- Jennifer Streeter
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.)
| | - Brandon M Schickling
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.)
| | - Shuxia Jiang
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.)
| | - Bojana Stanic
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.)
| | - William H Thiel
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.)
| | - Lokesh Gakhar
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.)
| | - Jon C D Houtman
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.)
| | - Francis J Miller
- From the Departments of Internal Medicine (B.M.S., S.J., B.S., W.H.T., F.J.M.), Microbiology (J.C.D.H.), Anatomy and Cell Biology (J.S.), Biochemistry (L.G.), and Protein Crystallography Facility (L.G.), University of Iowa, Iowa City; and Veterans Affair Medical Center, Iowa City, IA (F.J.M.).
| |
Collapse
|
41
|
Chan EC, Dusting GJ, Liu GS, Jiang F. Redox mechanisms of the beneficial effects of heme oxygenase in hypertension. J Hypertens 2014; 32:1379-86; discussion 1387. [DOI: 10.1097/hjh.0000000000000179] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
Abstract
SIGNIFICANCE Reactive oxygen species (ROS) play a critical role in vascular disease. While there are many possible sources of ROS, nicotinamide adenine dinucleotide phosphate (NADPH) oxidases play a central role. They are a source of "kindling radicals," which affect other enzymes, such as nitric oxide synthase endothelial nitric oxide synthase or xanthine oxidase. This is important, as risk factors for atherosclerosis (hypertension, diabetes, hypercholesterolemia, and smoking) regulate the expression and activity of NADPH oxidases in the vessel wall. RECENT ADVANCES There are seven isoforms in mammals: Nox1, Nox2, Nox3, Nox4, Nox5, Duox1 and Duox2. Nox1, Nox2, Nox4, and Nox5 are expressed in endothelium, vascular smooth muscle cells, fibroblasts, or perivascular adipocytes. Other homologues have not been found or are expressed at very low levels; their roles have not been established. Nox1/Nox2 promote the development of endothelial dysfunction, hypertension, and inflammation. Nox4 may have a role in protecting the vasculature during stress; however, when its activity is increased, it may be detrimental. Calcium-dependent Nox5 has been implicated in oxidative damage in human atherosclerosis. CRITICAL ISSUES NADPH oxidase-derived ROS play a role in vascular pathology as well as in the maintenance of normal physiological vascular function. We also discuss recently elucidated mechanisms such as the role of NADPH oxidases in vascular protection, vascular inflammation, pulmonary hypertension, tumor angiogenesis, and central nervous system regulation of vascular function and hypertension. FUTURE DIRECTIONS Understanding the role of individual oxidases and interactions between homologues in vascular disease is critical for efficient pharmacological regulation of vascular NADPH oxidases in both the laboratory and clinical practice.
Collapse
Affiliation(s)
- Anna Konior
- 1 Department of Internal Medicine, Jagiellonian University School of Medicine , Cracow, Poland
| | | | | | | |
Collapse
|
43
|
Bernard K, Hecker L, Luckhardt TR, Cheng G, Thannickal VJ. NADPH oxidases in lung health and disease. Antioxid Redox Signal 2014; 20:2838-53. [PMID: 24093231 PMCID: PMC4026303 DOI: 10.1089/ars.2013.5608] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
SIGNIFICANCE The evolution of the lungs and circulatory systems in vertebrates ensured the availability of molecular oxygen (O2; dioxygen) for aerobic cellular metabolism of internal organs in large animals. O2 serves as the physiologic terminal acceptor of mitochondrial electron transfer and of the NADPH oxidase (Nox) family of oxidoreductases to generate primarily water and reactive oxygen species (ROS), respectively. RECENT ADVANCES The purposeful generation of ROS by Nox family enzymes suggests important roles in normal physiology and adaptation, most notably in host defense against invading pathogens and in cellular signaling. CRITICAL ISSUES However, there is emerging evidence that, in the context of chronic stress and/or aging, Nox enzymes contribute to the pathogenesis of a number of lung diseases. FUTURE DIRECTIONS Here, we review evolving functions of Nox enzymes in normal lung physiology and emerging pathophysiologic roles in lung disease.
Collapse
Affiliation(s)
- Karen Bernard
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| | | | | | | | | |
Collapse
|
44
|
Park MT, Kim MJ, Suh Y, Kim RK, Kim H, Lim EJ, Yoo KC, Lee GH, Kim YH, Hwang SG, Yi JM, Lee SJ. Novel signaling axis for ROS generation during K-Ras-induced cellular transformation. Cell Death Differ 2014; 21:1185-97. [PMID: 24632950 DOI: 10.1038/cdd.2014.34] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 02/04/2014] [Accepted: 02/05/2014] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are well known to be involved in oncogene-mediated cellular transformation. However, the regulatory mechanisms underlying ROS generation in oncogene-transformed cells are unclear. In the present study, we found that oncogenic K-Ras induces ROS generation through activation of NADPH oxidase 1 (NOX1), which is a critical regulator for the K-Ras-induced cellular transformation. NOX1 was activated by K-Ras-dependent translocation of p47(phox), a subunit of NOX1 to plasma membrane. Of note, PKCδ, when it was activated by PDPK1, directly bound to the SH3-N domain of p47(phox) and catalyzed the phosphorylation on Ser348 and Ser473 residues of p47(phox) C-terminal in a K-Ras-dependent manner, finally leading to its membrane translocation. Notably, oncogenic K-Ras activated all MAPKs (JNK, ERK and p38); however, only p38 was involved in p47(phox)-NOX1-dependent ROS generation and consequent transformation. Importantly, K-Ras-induced activation of p38 led to an activation of PDPK1, which then signals through PKCδ, p47(phox) and NOX1. In agreement with the mechanism, inhibition of p38, PDPK1, PKCδ, p47(phox) or NOX1 effectively blocked K-Ras-induced ROS generation, anchorage-independent colony formation and tumor formation. Taken together, our findings demonstrated that oncogenic K-Ras activates the signaling cascade p38/PDPK1/PKCδ/p47(phox)/NOX1 for ROS generation and consequent malignant cellular transformation.
Collapse
Affiliation(s)
- M-T Park
- 1] Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea [2] Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, Korea
| | - M-J Kim
- 1] Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea [2] Low Dose Radiation Research Center, National Radiation Emergency Medical Science, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Korea
| | - Y Suh
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| | - R-K Kim
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| | - H Kim
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| | - E-J Lim
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| | - K-C Yoo
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| | - G-H Lee
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| | - Y-H Kim
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| | - S-G Hwang
- Division of Radiation Cancer Biology, Korea Institute of Radiological and Medical Sciences, Seoul 139-706, Korea
| | - J-M Yi
- Research Center, Dongnam Institute of Radiological and Medical Sciences, Busan, Korea
| | - S-J Lee
- Laboratory of Molecular Biochemistry, Department of Life Science, Research Institute for Natural Sciences, Hanyang University, Seoul 133-791, Korea
| |
Collapse
|
45
|
Smith SME, Min J, Ganesh T, Diebold B, Kawahara T, Zhu Y, McCoy J, Sun A, Snyder JP, Fu H, Du Y, Lewis I, Lambeth JD. Ebselen and congeners inhibit NADPH oxidase 2-dependent superoxide generation by interrupting the binding of regulatory subunits. ACTA ACUST UNITED AC 2014; 19:752-63. [PMID: 22726689 DOI: 10.1016/j.chembiol.2012.04.015] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2011] [Revised: 04/18/2012] [Accepted: 04/27/2012] [Indexed: 02/07/2023]
Abstract
NADPH oxidases (Nox) are a primary source of reactive oxygen species (ROS), which function in normal physiology and, when overproduced, in pathophysiology. Recent studies using mice deficient in Nox2 identify this isoform as a novel target against Nox2-implicated inflammatory diseases. Nox2 activation depends on the binding of the proline-rich domain of its heterodimeric partner p22phox to p47phox. A high-throughput screen that monitored this interaction via fluorescence polarization identified ebselen and several of its analogs as inhibitors. Medicinal chemistry was performed to explore structure-activity relationships and to optimize potency. Ebselen and analogs potently inhibited Nox1 and Nox2 activity but were less effective against other isoforms. Ebselen also blocked translocation of p47phox to neutrophil membranes. Thus, ebselen and its analogs represent a class of compounds that inhibit ROS generation by interrupting the assembly of Nox2-activating regulatory subunits.
Collapse
Affiliation(s)
- Susan M E Smith
- Department of Pathology, Emory School of Medicine, 615 Michael Street, Atlanta, GA 30322, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
The microbiota that populates the intestinal tract affects many physiological processes, such as cell proliferation, epithelial barrier function, and immune responses. However, the molecular mechanisms by which the microbiota influences these events remain unknown. It was recently reported by our research group that specific taxa of intestinal bacteria induce the rapid and transient enzymatic production of reactive oxygen species (ROS) within enterocytes. Whereas NADPH oxidase 2 (Nox2) catalyzed ROS generation in response to microbial perception by bone marrow-derived phagocytes is well-studied, the function of ROS generated by Nox1 in enterocytes in response to microbial signals is not fully understood. It is established that ROS can act as signaling molecules in diverse transduction pathways by the rapid and transient oxidation of oxidant-sensitive thiol groups harbored within sensor regulatory proteins. Because commensal-bacterial-stimulated ROS generation in enterocytes has been shown to induce a wide range of physiological processes, in our recent manuscript, we proposed a paradigm wherein the influence of the microbiota on intestinal physiology is mediated in part by redox-dependant signaling.
Collapse
|
47
|
Abstract
By definition, the mucosal immune system is responsible for interfacing with the outside world, specifically responding to external threats, of which pathogenic microbes represent a primary challenge. However, it has become apparent that the human host possesses a numerically vast and taxonomically diverse resident microbiota, predominantly in the gut, and also in the airway, genitourinary tract, and skin. The microbiota is generally considered symbiotic, and has been implicated in the regulation of cellular growth, restitution after injury, maintenance of barrier function, and importantly, in the induction, development, and modulation of immune responses. The mucosal immune system uses diverse mechanisms that protect the host from overt pathogens, but necessarily has coevolved to monitor, nurture, and exploit the normal microbiota. As a whole, mucosal immunity encompasses adaptive immune regulation that can involve systemic processes, local tissue-based innate and inflammatory events, intrinsic defenses, and highly conserved cell autonomous cytoprotective responses. Interestingly, specific taxa within the normal microbiota have been implicated in roles shaping specific adaptive, innate, and cell autonomous responses. Taken together, the normal microbiota exerts profound effects on the mucosal immune system, and likely plays key roles in human physiology and disease.
Collapse
Affiliation(s)
- Andrew S. Neish
- Department of Pathology, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
48
|
Ranayhossaini DJ, Rodriguez AI, Sahoo S, Chen BB, Mallampalli RK, Kelley EE, Csanyi G, Gladwin MT, Romero G, Pagano PJ. Selective recapitulation of conserved and nonconserved regions of putative NOXA1 protein activation domain confers isoform-specific inhibition of Nox1 oxidase and attenuation of endothelial cell migration. J Biol Chem 2013; 288:36437-50. [PMID: 24187133 PMCID: PMC3868757 DOI: 10.1074/jbc.m113.521344] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2013] [Indexed: 11/06/2022] Open
Abstract
Excessive vascular and colon epithelial reactive oxygen species production by NADPH oxidase isoform 1 (Nox1) has been implicated in a number of disease states, including hypertension, atherosclerosis, and neoplasia. A peptide that mimics a putative activation domain of the Nox1 activator subunit NOXA1 (NOXA1 docking sequence, also known as NoxA1ds) potently inhibited Nox1-derived superoxide anion (O2·-) production in a reconstituted Nox1 cell-free system, with no effect on Nox2-, Nox4-, Nox5-, or xanthine oxidase-derived reactive oxygen species production as measured by cytochrome c reduction, Amplex Red fluorescence, and electron paramagnetic resonance. The ability of NoxA1ds to cross the plasma membrane was tested by confocal microscopy in a human colon cancer cell line exclusively expressing Nox1 (HT-29) using FITC-labeled NoxA1ds. NoxA1ds significantly inhibited whole HT-29 carcinoma cell-derived O2·- generation. ELISA and fluorescence recovery after photobleaching experiments indicate that NoxA1ds, but not its scrambled control, binds Nox1. FRET experiments conducted using Nox1-YFP and NOXA1-CFP illustrate that NoxA1ds disrupts the binding interaction between Nox1 and NOXA1, whereas a control peptide did not. Moreover, hypoxia-induced human pulmonary artery endothelial cell O2·- production was completely inhibited by NoxA1ds. Human pulmonary artery endothelial cell migration under hypoxic conditions was also reduced by pretreatment with NoxA1ds. Our data indicate that a peptide recapitulating a putative activation subdomain of NOXA1 (NoxA1ds) is a highly efficacious and selective inhibitor of Nox1 activity and establishes a critical interaction site for Nox1-NOXA1 binding required for enzyme activation.
Collapse
Affiliation(s)
- Daniel J. Ranayhossaini
- From the Vascular Medicine Institute and
- Departments of Pharmacology and Chemical Biology and
| | - Andres I. Rodriguez
- From the Vascular Medicine Institute and
- Departments of Pharmacology and Chemical Biology and
| | | | - Beibei B. Chen
- Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261 and
| | - Rama K. Mallampalli
- Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261 and
- the Medical Specialty Service Line, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania 15240
| | - Eric E. Kelley
- From the Vascular Medicine Institute and
- Departments of Pharmacology and Chemical Biology and
| | - Gabor Csanyi
- From the Vascular Medicine Institute and
- Departments of Pharmacology and Chemical Biology and
| | - Mark T. Gladwin
- From the Vascular Medicine Institute and
- Pulmonary, Allergy, and Critical Care Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania 15261 and
| | | | - Patrick J. Pagano
- From the Vascular Medicine Institute and
- Departments of Pharmacology and Chemical Biology and
| |
Collapse
|
49
|
Woolley JF, Corcoran A, Groeger G, Landry WD, Cotter TG. Redox-regulated growth factor survival signaling. Antioxid Redox Signal 2013. [PMID: 23198948 DOI: 10.1089/ars.2012.5028] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Once the thought of as unwanted byproducts of cellular respiration in eukaryotes, reactive oxygen species (ROS) have been shown to facilitate essential physiological roles. It is now understood that ROS are critical mediators of intracellular signaling. Control of signal transduction downstream of growth factor receptors by ROS is a complex process whose details are only recently coming to light. RECENT ADVANCES Indeed, recent evidence points to control of signal propagation by ROS at multiple levels in the typical cascade. Growth factor stimulation activates nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Noxs) at the membrane, producing superoxide in the extracellular matrix, which is catalyzed to the membrane-permeable hydrogen peroxide (H2O2) that mediates intracellular signaling events. CRITICAL ISSUES The potential for H2O2, however, to disrupt cellular functions by damaging proteins and nucleic acids demands that its levels are kept in check by receptor-associated peroxiredoxins. This interplay of Nox and peroxiredoxin activity moderates levels of H2O2 sufficiently to modify signaling partners locally. Among the best studied of these partners are redox-controlled phosphatases that are inactivated by H2O2. Phosphatases regulate signal propagation downstream of receptors, and thus their inactivation allows a further level of control. Transmission of information further downstream to targets such as transcription factors, themselves regulated by ROS, completes this pathway. FUTURE DIRECTIONS Thus, signal propagation or attenuation can be dictated by ROS at multiple points. Given the complex nature of these processes, we envisage the emerging trends in the field of redox signaling in the context of growth factor stimulation.
Collapse
Affiliation(s)
- John F Woolley
- Tumour Biology Laboratory, Biochemistry Department, Bioscience Research Institute, University College , Cork, Ireland
| | | | | | | | | |
Collapse
|
50
|
Abstract
The microbiota that occupies the mammalian intestine can modulate a range of physiological functions, including control over immune responses, epithelial barrier function, and cellular proliferation. While commensal prokaryotic organisms are well known to stimulate inflammatory signaling networks, less is known about control over homeostatic pathways. Recent work has shown that gut epithelia contacted by enteric commensal bacteria rapidly generate reactive oxygen species (ROS). While the induced production of ROS in professional phagocytes via stimulation of formyl peptide receptors (FPRs) and activation of NADPH oxidase 2 (Nox2) is a well-studied process, ROS are also similarly elicited in other cell types, including intestinal epithelia, in response to microbial signals via FPRs and the epithelial NADPH oxidase 1 (Nox1). ROS generated by Nox enzymes have been shown to function as critical second messengers in multiple signal transduction pathways via the rapid and transient oxidative inactivation of a distinct class of sensor proteins bearing oxidant-sensitive thiol groups. These redox-sensitive proteins include tyrosine phosphatases that serve as regulators of MAP kinase pathways, focal adhesion kinase, as well as components involved in NF-κB activation. As microbe-elicited ROS has been shown to stimulate cellular proliferation and motility, and to modulate innate immune signaling, we hypothesize that many of the established effects of the normal microbiota on intestinal physiology may be at least partially mediated by this ROS-dependent mechanism.
Collapse
Affiliation(s)
- Andrew S Neish
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine , Atlanta, GA , USA
| |
Collapse
|