1
|
Zmudzka M, Szramel J, Karasinski J, Nieckarz Z, Zoladz JA, Majerczak J. Physical activity reverses the aging induced decline in angiogenic potential in the fast locomotory muscles of mice. Sci Rep 2025; 15:8848. [PMID: 40087472 PMCID: PMC11909166 DOI: 10.1038/s41598-025-93176-1] [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: 11/22/2024] [Accepted: 03/05/2025] [Indexed: 03/17/2025] Open
Abstract
Fast locomotory muscles, which are responsible for generating the highest power outputs, are more vulnerable to aging than slow muscles. In this study, we aimed to evaluate the impact of middle age and voluntary physical activity on capillarization and angiogenic potential in fast locomotory muscles. Middle-aged (M-group) and young (Y-group) wild-type FVB female mice were randomly assigned to either the sedentary or trained group undergoing 8-week spontaneous wheel running (8-sWR). Capillary density (assessed via immunohistochemical capillary staining and Western immunoblotting) of the fast locomotory muscles in the M-group (15-months old) was not significantly different compared to the Y-group (4-months old). Nevertheless, the expression of key pro-angiogenic genes in the fast muscle of the M-group was lower than that in the fast muscle of Y-group. 8-sWR had no impact on muscle capillarization; however, it increased fast muscle Vegfa expression in both the M and Y groups. We concluded that although fast muscle capillarization is still preserved in middle age, nevertheless the angiogenic potential (at least at the level of gene expression) is significantly reduced at this stage of aging. Moderate-intensity voluntary physical activity had no effect on capillary density, but it increased the angiogenic potential of the fast muscle.
Collapse
Affiliation(s)
- Magdalena Zmudzka
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, Skawinska 8 Street, Krakow, 31-066, Poland
| | - Joanna Szramel
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, Skawinska 8 Street, Krakow, 31-066, Poland
| | - Janusz Karasinski
- Department of Cell Biology and Imaging, Institute of Zoology and Biomedical Research, Faculty of Biology, Jagiellonian University, Krakow, Poland
| | - Zenon Nieckarz
- Department of Experimental Computer Physics, Marian Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Jerzy A Zoladz
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, Skawinska 8 Street, Krakow, 31-066, Poland
| | - Joanna Majerczak
- Chair of Exercise Physiology and Muscle Bioenergetics, Faculty of Health Sciences, Jagiellonian University Medical College, Skawinska 8 Street, Krakow, 31-066, Poland.
| |
Collapse
|
2
|
Ebding J, Mazzone F, Kins S, Pielage J, Maritzen T. How neurons cope with oxidative stress. Biol Chem 2025:hsz-2024-0146. [PMID: 39988910 DOI: 10.1515/hsz-2024-0146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/31/2025] [Indexed: 02/25/2025]
Abstract
Neurons are highly dependent on mitochondrial respiration for energy, rendering them vulnerable to oxidative stress. Reactive oxygen species (ROS), by-products of oxidative phosphorylation, can damage lipids, proteins, and DNA, potentially triggering cell death pathways. This review explores the neuronal vulnerability to ROS, highlighting metabolic adaptations and antioxidant systems that mitigate oxidative damage. Balancing metabolic needs and oxidative stress defenses is critical for neurons, as disruptions are implicated in neurodegenerative diseases. Neurons uniquely modulate metabolic pathways, favoring glycolysis over oxidative phosphorylation in cell bodies, to minimize harmful ROS production. Key antioxidants, including superoxide dismutases and glutathione peroxidases, play crucial roles in neuronal protection, as evident from genetic studies linking deficiencies to neurodegeneration. Notably, neurons have the ability to adapt to oxidative conditions in compartment-specific manners and also utilize ROS as a signaling molecule to promote adaptive synaptic plasticity. Future research should aim to elucidate differential ROS signaling and antioxidant responses across neuronal compartments for improved therapeutic strategies.
Collapse
Affiliation(s)
- Johannes Ebding
- Department for Neurobiology and Zoology, 2026562 RPTU University Kaiserslautern-Landau, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| | - Fiorella Mazzone
- Department for Nanophysiology, 2026562 RPTU University Kaiserslautern-Landau, Paul-Ehrlich-Straße 23, D-67663 Kaiserslautern, Germany
| | - Stefan Kins
- Department for Human Biology, 2026562 RPTU University Kaiserslautern-Landau, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| | - Jan Pielage
- Department for Neurobiology and Zoology, 2026562 RPTU University Kaiserslautern-Landau, Erwin-Schrödinger-Straße 13, D-67663 Kaiserslautern, Germany
| | - Tanja Maritzen
- Department for Nanophysiology, 2026562 RPTU University Kaiserslautern-Landau, Paul-Ehrlich-Straße 23, D-67663 Kaiserslautern, Germany
| |
Collapse
|
3
|
Varone M, Scavo G, Colardo M, Martella N, Pensabene D, Bisesto E, Del Busso A, Segatto M. p75NTR Modulation Reduces Oxidative Stress and the Expression of Pro-Inflammatory Mediators in a Cell Model of Rett Syndrome. Biomedicines 2024; 12:2624. [PMID: 39595188 PMCID: PMC11592079 DOI: 10.3390/biomedicines12112624] [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: 10/19/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Rett syndrome (RTT) is an early-onset neurological disorder primarily affecting females, leading to severe cognitive and physical disabilities. Recent studies indicate that an imbalance of redox homeostasis and exacerbated inflammatory responses are key players in the clinical manifestations of the disease. Emerging evidence highlights that the p75 neurotrophin receptor (p75NTR) is implicated in the regulation of oxidative stress (OS) and inflammation. Thus, this study is aimed at investigating the effects of p75NTR modulation by LM11A-31 on fibroblasts derived from RTT donors. Methods: RTT cells were treated with 0.1 µM of LM11A-31 for 24 h, and results were obtained using qPCR, immunofluorescence, ELISA, and Western blot techniques. Results: Our findings demonstrate that LM11A-31 reduces OS markers in RTT fibroblasts. Specifically, p75NTR modulation by LM11A-31 restores protein glutathionylation and reduces the expression of the pro-oxidant enzyme NOX4. Additionally, LM11A-31 significantly decreases the expression of the pro-inflammatory mediators interleukin-6 and interleukin-8. Additionally, LM11A-31 normalizes the expression levels of transcription factors involved in the regulation of the antioxidant response and inflammation. Conclusions: Collectively, these data suggest that p75NTR modulation may represent an effective therapeutic target to improve redox balance and reduce inflammation in RTT.
Collapse
Affiliation(s)
- Michela Varone
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
| | - Giuseppe Scavo
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
| | - Mayra Colardo
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
| | - Noemi Martella
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
| | - Daniele Pensabene
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
- Department of Science, University Roma Tre, 00146 Rome, Italy
| | - Emanuele Bisesto
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
| | - Andrea Del Busso
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
| | - Marco Segatto
- Department of Biosciences and Territory, University of Molise, Contrada Fonte Lappone, 86090 Pesche, Italy; (M.V.); (G.S.); (M.C.); (N.M.); (D.P.); (E.B.); (A.D.B.)
| |
Collapse
|
4
|
Barra W, Queiroz B, Perez A, Romero T, Ferreira R, Duarte I. Study on peripheral antinociception induced by hydrogen peroxide (H 2O 2): characterization and mechanisms. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:7927-7938. [PMID: 38753048 DOI: 10.1007/s00210-024-03087-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/05/2024] [Indexed: 10/04/2024]
Abstract
The present study aimed to evaluate the possible peripheral H2O2-induced antinociception and determine the involvement of opioidergic, cannabinoidergic and nitrergic systems, besides potassium channels in its antinociceptive effect. Prostaglandin E2 was used to induce hyperalgesia in male Swiss mice using the mechanical paw pressure test. H2O2 (0.1, 0.2, 0.3 µg/paw) promoted a dose-dependent antinociceptive effect that was not observed in contralateral paw. Female mice also showed antinociception in the model. The partial H2O2-induced antinociception was potentiated by the inhibitor of catalase enzyme, aminotriazole (40, 60, 80 µg/paw). The antinociception was not reversed by opioid and cannabinoid receptor antagonists naloxone, AM 251 and AM 630. The involvement of nitric oxide (NO) was observed by the reversal of H2O2-induced antinociception using the non-selective inhibitor of nitric oxide synthases L-NOarg and by inhibition of iNOS (L-NIL), eNOS (L-NIO) and nNOS (L-NPA). ODQ, a cGMP-forming enzyme selective inhibitor, also reversed the antinociception. The blockers of potassium channels voltage-gated (TEA), ATP-sensitive (glibenclamide), large (paxillin) and small (dequalinium) conductance calcium-activated were able to revert H2O2 antinociception. Our data suggest that H2O2 induced a peripheral antinociception in mice and the NO pathway and potassium channels (voltage-gated, ATP-sensitive, calcium-activated) are involved in this mechanism. However, the role of the opioid and cannabinoid systems was not evidenced.
Collapse
Affiliation(s)
- Walace Barra
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, City Belo Horizonte, Brazil
| | - Bárbara Queiroz
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, City Belo Horizonte, Brazil
| | - Andrea Perez
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, City Belo Horizonte, Brazil
| | - Thiago Romero
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, City Belo Horizonte, Brazil
| | - Renata Ferreira
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, City Belo Horizonte, Brazil
| | - Igor Duarte
- Laboratory of Pain and Analgesia, Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, City Belo Horizonte, Brazil.
| |
Collapse
|
5
|
Long S, Zheng Y, Deng X, Guo J, Xu Z, Scharffetter-Kochanek K, Dou Y, Jiang M. Maintaining mitochondrial DNA copy number mitigates ROS-induced oocyte decline and female reproductive aging. Commun Biol 2024; 7:1229. [PMID: 39354016 PMCID: PMC11445474 DOI: 10.1038/s42003-024-06888-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 09/12/2024] [Indexed: 10/03/2024] Open
Abstract
Oocytes play a crucial role in transmitting maternal mitochondrial DNA (mtDNA), essential for the continuation of species. However, the effects of mitochondrial reactive oxygen species (ROS) on mammalian oocyte maturation and mtDNA maintenance remain unclear. We investigated this by conditionally knocking out the Sod2 gene in primordial follicles, elevating mitochondrial matrix ROS levels from early oocyte stages. Our data indicates that reproductive aging in Sod2 conditional knockout females begins at 6 months, with oxidative stress impairing oocyte quality, particularly affecting OXPHOS complex II and mtDNA-encoded mRNA levels. Despite unchanged mtDNA mutation load, mtDNA copy numbers exhibited significant variations. Strikingly, reducing mtDNA copy numbers by reducing mtSSB protein, crucial for mtDNA replication, accelerated reproductive aging onset to three months, underscoring the critical role of mtDNA copy number maintenance under oxidative stress conditions. This research provides new insights into the relationship among mitochondrial ROS, mtDNA, and reproductive aging, offering potential strategies for delaying aging-related fertility decline.
Collapse
Affiliation(s)
- Shiyun Long
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Yunchao Zheng
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Xiaoling Deng
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
- Fudan University, Shanghai, China
| | - Jing Guo
- NHC Key Laboratory of Human Stem Cell and Reproductive Engineering, School of Basic Medical Sciences, Central South University, Changsha, Hunan, China
| | - Zhe Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Karin Scharffetter-Kochanek
- Klinik für Dermatologie und Allergologie, Universitätsklinikum Ulm, Albert-Einstein-Allee 23, 89081, Ulm, Deutschland
| | - Yanmei Dou
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| | - Min Jiang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Rody E, Zwaig J, Derish I, Khan K, Kachurina N, Gendron N, Giannetti N, Schwertani A, Cecere R. Evaluating the Reparative Potential of Secretome from Patient-Derived Induced Pluripotent Stem Cells during Ischemia-Reperfusion Injury in Human Cardiomyocytes. Int J Mol Sci 2024; 25:10279. [PMID: 39408608 PMCID: PMC11477076 DOI: 10.3390/ijms251910279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/03/2024] [Indexed: 10/20/2024] Open
Abstract
During a heart attack, ischemia causes losses of billions of cells; this is especially concerning given the minimal regenerative capability of cardiomyocytes (CMs). Heart remuscularization utilizing stem cells has improved cardiac outcomes despite little cell engraftment, thereby shifting focus to cell-free therapies. Consequently, we chose induced pluripotent stem cells (iPSCs) given their pluripotent nature, efficacy in previous studies, and easy obtainability from minimally invasive techniques. Nonetheless, using iPSC secretome-based therapies for treating injured CMs in a clinical setting is ill-understood. We hypothesized that the iPSC secretome, regardless of donor health, would improve cardiovascular outcomes in the CM model of ischemia-reperfusion (IR) injury. Episomal-generated iPSCs from healthy and dilated cardiomyopathy (DCM) donors, passaged 6-10 times, underwent 24 h incubation in serum-free media. Protein content of the secretome was analyzed by mass spectroscopy and used to treat AC16 immortalized CMs during 5 h reperfusion following 24 h of hypoxia. IPSC-derived secretome content, independent of donor health status, had elevated expression of proteins involved in cell survival pathways. In IR conditions, iPSC-derived secretome increased cell survival as measured by metabolic activity (p < 0.05), cell viability (p < 0.001), and maladaptive cellular remodelling (p = 0.052). Healthy donor-derived secretome contained increased expression of proteins related to calcium contractility compared to DCM donors. Congruently, only healthy donor-derived secretomes improved CM intracellular calcium concentrations (p < 0.01). Heretofore, secretome studies mainly investigated differences relating to cell type rather than donor health. Our work suggests that healthy donors provide more efficacious iPSC-derived secretome compared to DCM donors in the context of IR injury in human CMs. These findings illustrate that the regenerative potential of the iPSC secretome varies due to donor-specific differences.
Collapse
Affiliation(s)
- Elise Rody
- Department of Surgery, Division of Cardiac Surgery, McGill University Health Center, Montreal, QC H4A 3J1, Canada
| | - Jeremy Zwaig
- Faculty of Medicine, McGill University, Montreal, QC H3G 2M1, Canada; (J.Z.)
| | - Ida Derish
- Faculty of Medicine, McGill University, Montreal, QC H3G 2M1, Canada; (J.Z.)
- Department of Surgical and Interventional Sciences, McGill University, Montreal, QC H3G 1A4, Canada
| | - Kashif Khan
- Faculty of Medicine, McGill University, Montreal, QC H3G 2M1, Canada; (J.Z.)
- Department of Medicine, Division of Cardiology, McGill University Health Center, Montreal, QC H4A 3J1, Canada (N.G.)
| | - Nadezda Kachurina
- Department of Medicine, Division of Cardiology, McGill University Health Center, Montreal, QC H4A 3J1, Canada (N.G.)
| | - Natalie Gendron
- Department of Medicine, Division of Cardiology, McGill University Health Center, Montreal, QC H4A 3J1, Canada (N.G.)
| | - Nadia Giannetti
- Department of Medicine, Division of Cardiology, McGill University Health Center, Montreal, QC H4A 3J1, Canada (N.G.)
| | - Adel Schwertani
- Department of Medicine, Division of Cardiology, McGill University Health Center, Montreal, QC H4A 3J1, Canada (N.G.)
| | - Renzo Cecere
- Department of Surgery, Division of Cardiac Surgery, McGill University Health Center, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
7
|
Gogna T, Housden BE, Houldsworth A. Exploring the Role of Reactive Oxygen Species in the Pathogenesis and Pathophysiology of Alzheimer's and Parkinson's Disease and the Efficacy of Antioxidant Treatment. Antioxidants (Basel) 2024; 13:1138. [PMID: 39334797 PMCID: PMC11429442 DOI: 10.3390/antiox13091138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/30/2024] Open
Abstract
Alzheimer's (AD) and Parkinson's Disease (PD) are life-altering diseases that are characterised by progressive memory loss and motor dysfunction. The prevalence of AD and PD is predicted to continuously increase. Symptoms of AD and PD are primarily mediated by progressive neuron death and dysfunction in the hippocampus and substantia nigra. Central features that drive neurodegeneration are caspase activation, DNA fragmentation, lipid peroxidation, protein carbonylation, amyloid-β, and/or α-synuclein formation. Reactive oxygen species (ROS) increase these central features. Currently, there are limited therapeutic options targeting these mechanisms. Antioxidants reduce ROS levels by the induction of antioxidant proteins and direct neutralisation of ROS. This review aims to assess the effectiveness of antioxidants in reducing ROS and neurodegeneration. Antioxidants enhance major endogenous defences against ROS including superoxide dismutase, catalase, and glutathione. Direct neutralisation of ROS by antioxidants protects against ROS-induced cytotoxicity. The combination of Indirect and direct protective mechanisms prevents ROS-induced α-synuclein and/or amyloid-β formation. Antioxidants ameliorate ROS-mediated oxidative stress and subsequent deleterious downstream effects that promote apoptosis. As a result, downstream harmful events including neuron death, dysfunction, and protein aggregation are decreased. The protective effects of antioxidants in human models have yet to directly replicate the success seen in cell and animal models. However, the lack of diversity in antioxidants for clinical trials prevents a definitive answer if antioxidants are protective. Taken together, antioxidant treatment is a promising avenue in neurodegenerative disease therapy and subsequent clinical trials are needed to provide a definitive answer on the protective effects of antioxidants. No current treatment strategies have significant impact in treating advanced AD and PD, but new mimetics of endogenous mitochondrial antioxidant enzymes (Avasopasem Manganese, GC4419 AVA) may be a promising innovative option for decelerating neurodegenerative progress in the future at the mitochondrial level of OS.
Collapse
Affiliation(s)
- Talin Gogna
- Neuroscience, Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| | - Benjamin E Housden
- Living Systems Institute, Clinical and Biomedical Sciences, University of Exeter, Stocker Road, Exeter EX4 4QD, UK
| | - Annwyne Houldsworth
- Clinical and Biomedical Sciences, University of Exeter Medical School, Exeter EX2 4TH, UK
| |
Collapse
|
8
|
Ulfig A, Jakob U. Cellular oxidants and the proteostasis network: balance between activation and destruction. Trends Biochem Sci 2024; 49:761-774. [PMID: 39168791 PMCID: PMC11731897 DOI: 10.1016/j.tibs.2024.07.001] [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: 04/05/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 08/23/2024]
Abstract
Loss of protein homeostasis (proteostasis) is a common hallmark of aging and age-associated diseases. Considered as the guardian of proteostasis, the proteostasis network (PN) acts to preserve the functionality of proteins during their lifetime. However, its activity declines with age, leading to disease manifestation. While reactive oxygen species (ROS) were traditionally considered culprits in this process, recent research challenges this view. While harmful at high concentrations, moderate ROS levels protect the cell against age-mediated onset of proteotoxicity by activating molecular chaperones, stress response pathways, and autophagy. This review explores the nuanced roles of ROS in proteostasis and discusses the most recent findings regarding the redox regulation of the PN and its potential in extending healthspan and delaying age-related pathologies.
Collapse
Affiliation(s)
- Agnes Ulfig
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Ursula Jakob
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA; Biological Chemistry Department, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
9
|
Stovall K, Patel M, Franklin JL. The intrinsic apoptotic pathway lies upstream of reactive species production in cortical neurons and age-related oxidative stress in the brain. Mol Cell Neurosci 2023; 127:103901. [PMID: 37729979 DOI: 10.1016/j.mcn.2023.103901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 09/11/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023] Open
Abstract
A BAX- and mitochondria-dependent production of reactive oxygen species (ROS) and reactive species (reactive nitrogen species, RNS) lying downstream of these ROS occurs in apoptotic and nonapoptotic mouse sympathetic neurons and cerebellar granule cells in cell culture. These ROS have been shown to lie downstream of caspase 3 in mouse sympathetic neurons. Here we show that BAX is necessary for similar ROS production in apoptotic and nonapoptotic mouse cortical neurons in cell culture and that it also positively regulates oxidative stress in the brains of mice of different ages. Brains from mice with genetically reduced levels of mitochondrial superoxide dismutase 2 (SOD2) exhibited elevated levels of DNA strand breaks consistent with oxidative damage. Lipid peroxides were also elevated at some ages in comparison to the brains of wild type animals. BAX deletion in these mice reduced both brain DNA strand breaks and lipid peroxide levels to well below those of wild type animals. Deletion of caspase 3 greatly reduced age-augmented levels of brain oxidative stress markers including lipid peroxides, oxidized DNA, and nitrosylated proteins. These findings indicate that BAX contributes to ROS production in mouse cortical neurons, to oxidative stress their brains, and that this effect is likely mediated via caspase 3 activity.
Collapse
Affiliation(s)
- Kyndra Stovall
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, 357 Wilson Pharmacy, Athens, GA 30602, USA.
| | - Mital Patel
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, 357 Wilson Pharmacy, Athens, GA 30602, USA
| | - James L Franklin
- Department of Pharmaceutical and Biomedical Sciences, The University of Georgia College of Pharmacy, 357 Wilson Pharmacy, Athens, GA 30602, USA.
| |
Collapse
|
10
|
Yang X, Zhang Z, Shen X, Xu J, Weng Y, Wang W, Xue J. Clostridium butyricum and its metabolite butyrate promote ferroptosis susceptibility in pancreatic ductal adenocarcinoma. Cell Oncol (Dordr) 2023; 46:1645-1658. [PMID: 37261698 DOI: 10.1007/s13402-023-00831-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2023] [Indexed: 06/02/2023] Open
Abstract
PURPOSE Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with limited therapeutic options. The diversity and composition of the intratumoral microbiota are associated with PDAC outcomes, and modulating the tumor microbiota has the potential to influence tumor growth and the host immune response. Here, we explore whether intervention with butyrate-producing probiotics can limit PDAC progression. METHODS Based on the TCGA (PAAD) database, we analyzed the differential communities of intratumoral microbiota in PDAC patients with long survival and short survival and explored the relevant mechanisms of Clostridium butyricum and its metabolite butyrate in the treatment of PDAC. Treatment with Clostridium butyricum or butyrate in combination with the ferroptosis inducer RSL3 in a PDAC mouse model has an inhibitory effect on PDAC progression. The potential molecular mechanisms were verified by flow cytometry, RNA-seq, Western blotting, qRT‒PCR and immunofluorescence. RESULTS We found that the tumoral butyrate-producing microbiota was linked to a better prognosis and less aggressive features of PDAC. Intervention with Clostridium butyricum or its metabolite butyrate triggered superoxidative stress and intracellular lipid accumulation, which enhanced ferroptosis susceptibility in PDAC. CONCLUSION Our study reveals a novel antitumor mechanism of butyrate and suggests the therapeutic potential of butyrate-producing probiotics in PDAC.
Collapse
Affiliation(s)
- Xiaotong Yang
- State Key Laboratory of Systems Medicine for Cancer , Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, 160 Pujian Rd, Shanghai, 200127, China
| | - Zhengyan Zhang
- State Key Laboratory of Systems Medicine for Cancer , Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, 160 Pujian Rd, Shanghai, 200127, China
| | - Xuqing Shen
- State Key Laboratory of Systems Medicine for Cancer , Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, 160 Pujian Rd, Shanghai, 200127, China
| | - Junyi Xu
- State Key Laboratory of Systems Medicine for Cancer , Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, 160 Pujian Rd, Shanghai, 200127, China
| | - Yawen Weng
- State Key Laboratory of Systems Medicine for Cancer , Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, 160 Pujian Rd, Shanghai, 200127, China
| | - Wei Wang
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, No 100, Haining Road, Shanghai, 200080, China.
| | - Jing Xue
- State Key Laboratory of Systems Medicine for Cancer , Stem Cell Research Center, Ren Ji Hospital, Shanghai Cancer Institute, Shanghai Jiao Tong University School of Medicine, 160 Pujian Rd, Shanghai, 200127, China.
| |
Collapse
|
11
|
Kim M, Hong T, An G, Lim W, Song G. Toxic effects of benfluralin on zebrafish embryogenesis via the accumulation of reactive oxygen species and apoptosis. Comp Biochem Physiol C Toxicol Pharmacol 2023; 273:109722. [PMID: 37597713 DOI: 10.1016/j.cbpc.2023.109722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 08/13/2023] [Accepted: 08/16/2023] [Indexed: 08/21/2023]
Abstract
The dinitroaniline herbicide benfluralin is used weed control in conventional systems and poses a high risk of accumulation in aquatic systems. Previous studies have shown the toxic effects of benfluralin on non-target organisms; however, its developmental toxicity in vertebrates has not yet been reported. This study demonstrated the developmental toxicity of benfluralin and its mechanism of action, using zebrafish as an aquatic vertebrate model. Benfluralin induces morphological and physiological alterations in body length, yolk sac, and heart edema. We also demonstrated a reactive oxygen species (ROS) increase of approximately 325.53 % compared with the control group after 20 μM benfluralin-treatment. In addition, the malformation of the heart and vascular structures was identified using transgenic flk1:eGFP zebrafish models at 20 μM concentration benfluralin exposure. Moreover, benfluralin induced small livers, approximately 59.81 % of normal liver size, via abnormal development of the liver as observed in the transgenic L-fabp:dsRed zebrafish. Benfluralin also inhibits normal growth via abnormal expression of cell cycle regulatory genes and increases oxidative stress, inflammation, and apoptosis. Collectively, we elucidated the mechanisms associated with benfluralin toxicity, which lead to various abnormalities and developmental toxicities in zebrafish. Therefore, this study provides information on the parameters used to assess developmental toxicity in other aquatic organisms, such as herbicides, pesticides, and environmental contaminants.
Collapse
Affiliation(s)
- Miji Kim
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Taeyeon Hong
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Garam An
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea
| | - Whasun Lim
- Department of Biological Sciences, Sungkyunkwan University, Suwon 16419, Republic of Korea.
| | - Gwonhwa Song
- Institute of Animal Molecular Biotechnology and Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
12
|
Wang G, Laranjeiro R, LeValley S, Van Raamsdonk JM, Driscoll M. The glyoxylate shunt protein ICL-1 protects from mitochondrial superoxide stress through activation of the mitochondrial unfolded protein response. Free Radic Biol Med 2023; 208:771-779. [PMID: 37758122 DOI: 10.1016/j.freeradbiomed.2023.09.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/05/2023] [Accepted: 09/24/2023] [Indexed: 10/03/2023]
Abstract
Disrupting mitochondrial superoxide dismutase (SOD) causes neonatal lethality in mice and death of flies within 24 h after eclosion. Deletion of mitochondrial sod genes in C. elegans impairs fertility as well, but surprisingly is not detrimental to survival of progeny generated. The comparison of metabolic pathways among mouse, flies and nematodes reveals that mice and flies lack the glyoxylate shunt, a shortcut that bypasses part of the tricarboxylic acid (TCA) cycle. Here we show that ICL-1, the sole protein that catalyzes the glyoxylate shunt, is critical for protection against embryonic lethality resulting from elevated levels of mitochondrial superoxide. In exploring the mechanism by which ICL-1 protects against ROS-mediated embryonic lethality, we find that ICL-1 is required for the efficient activation of mitochondrial unfolded protein response (UPRmt) and that ATFS-1, a key UPRmt transcription factor and an activator of icl-1 gene expression, is essential to limit embryonic/neonatal lethality in animals lacking mitochondrial SOD. In sum, we identify a biochemical pathway that highlights a molecular strategy for combating toxic mitochondrial superoxide consequences in cells.
Collapse
Affiliation(s)
- Guoqiang Wang
- Department of Molecular Biology and Biochemistry, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Ricardo Laranjeiro
- Department of Molecular Biology and Biochemistry, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA
| | - Stephanie LeValley
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Jeremy M Van Raamsdonk
- Laboratory of Aging and Neurodegenerative Disease, Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA; Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Monica Driscoll
- Department of Molecular Biology and Biochemistry, School of Arts and Sciences, Rutgers, the State University of New Jersey, Piscataway, NJ, USA.
| |
Collapse
|
13
|
Solanki N, Bhavsar S. Neuroprotective effects of Bauhinia variegata in ameliorating diabetic neuropathy and neurodegeneration. Am J Transl Res 2023; 15:6148-6158. [PMID: 37969205 PMCID: PMC10641339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/27/2023] [Indexed: 11/17/2023]
Abstract
INTRODUCTION In diabetic neuropathy with neurodegeneration (DNN), a serious diabetes consequence, extreme hyperglycemia destroys neurons in the brain and limbs. The main therapies for this condition are glucose control and pain management. Phytopharmacology is thought to be more successful in addressing the pain and blood sugar management issues associated with DNN. The objective of this study was to investigate how Bauhinia variegata (BV) could offer protection against streptozotocin (STZ)-induced diabetic neuropathy. METHODOLOGY STZ-associated DNN was induced in rats, and these diabetic rats were treated with BV at 200 mg/kg and 400 mg/kg doses for 28 days. Blood glucose (BG), serum nitrite, lipid peroxidation, antioxidants, C-reactive protein, behavioral, and histopathological parameters were assessed. RESULTS BV dramatically reduced BG and HbA1c levels in diabetic rats, according to the findings. The levels of superoxide dismutase and catalase both rose significantly. Both lipid peroxidation and serum nitrite levels were drastically decreased with BV treatment. In this study, it was found that BV has anti-hyperglycemic and anti-inflammatory effects on DNN. This was shown by a significant drop in C-reactive protein in diabetic rats, which was a key factor in diabetic neuropathy. Thermal hyperalgesia was significantly alleviated after BV therapy, and diabetic rats' pain thresholds improved. CONCLUSION Present study concluded that BV treatment has excellent glycemic control, a high antioxidant status, and relevant pain-relieving potential in diabetic neuropathy with neurodegeneration by reversing thermal hyperalgesia and decreasing hypeglycemia in diabetic rats.
Collapse
Affiliation(s)
- Nilay Solanki
- Department of Pharmacology, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT CampusChanga 388421, Gujarat, India
| | - Shailesh Bhavsar
- Department of Veterinary Pharmacology and Toxicology, Kamdhenu UniversityAnand, Gujarat, India
| |
Collapse
|
14
|
Gupta SV, Campos L, Schmidt KH. Mitochondrial superoxide dismutase Sod2 suppresses nuclear genome instability during oxidative stress. Genetics 2023; 225:iyad147. [PMID: 37638880 PMCID: PMC10550321 DOI: 10.1093/genetics/iyad147] [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: 02/24/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Oxidative stress can damage DNA and thereby contribute to genome instability. To avoid an imbalance or overaccumulation of reactive oxygen species (ROS), cells are equipped with antioxidant enzymes that scavenge excess ROS. Cells lacking the RecQ-family DNA helicase Sgs1, which contributes to homology-dependent DNA break repair and chromosome stability, are known to accumulate ROS, but the origin and consequences of this oxidative stress phenotype are not fully understood. Here, we show that the sgs1 mutant exhibits elevated mitochondrial superoxide, increased mitochondrial mass, and accumulation of recombinogenic DNA lesions that can be suppressed by antioxidants. Increased mitochondrial mass in the sgs1Δ mutant is accompanied by increased mitochondrial branching, which was also inducible in wildtype cells by replication stress. Superoxide dismutase Sod2 genetically interacts with Sgs1 in the suppression of nuclear chromosomal rearrangements under paraquat (PQ)-induced oxidative stress. PQ-induced chromosome rearrangements in the absence of Sod2 are promoted by Rad51 recombinase and the polymerase subunit Pol32. Finally, the dependence of chromosomal rearrangements on the Rev1/Pol ζ mutasome suggests that under oxidative stress successful DNA synthesis during DNA break repair depends on translesion DNA synthesis.
Collapse
Affiliation(s)
- Sonia Vidushi Gupta
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Lillian Campos
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
| | - Kristina Hildegard Schmidt
- Department of Molecular Biosciences, University of South Florida, 4202 East Fowler Avenue, Tampa, FL 33620, USA
- Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
15
|
Rabinovitz R, Eynan M. CNS-oxygen toxicity and blood glucose levels in MnSOD enzyme knockdown mice. Respir Physiol Neurobiol 2023; 316:104122. [PMID: 37481014 DOI: 10.1016/j.resp.2023.104122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 07/04/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023]
Abstract
Many studies have been conducted in the search for the mechanism underlying CNS-oxygen toxicity (OT), which may be fatal when diving with a closed-circuit apparatus. We investigated the influence of hyperbaric oxygen (HBO) on blood glucose level (BGL) in Mn-superoxide dismutase (SOD2) knockdown mice regarding CNS-OT in particular under stress conditions such as hypoglycemia or hyperglycemia. Two groups of mice were used: SOD2 knockdown (Heterozygous, HET) mice and their WT family littermates. Animals were exposed to HBO from 2 up to 5 atmosphere absolute (ATA). Blood samples were drawn before and after each exposure for measurement of BGL. The mice were sacrificed following the final exposure, which was at 5 ATA. We used RT-PCR and Western blot to measure levels of glucose transporter 1 (GLUT1) and hypoxia inducible factor (HIF)1a in the cortex and hippocampus. In the hypoglycemic condition, the HET mice were more sensitive to oxidative stress than the WT. In addition, following exposure to sub-toxic HBO, which does not induce CNS-OT, BGL were higher in the HET mice compared with the WT. The expression of mRNA of GLUT1 and HIF-1a decreased in the hippocampus in the HET mice, while the protein level decreased in the HET and WT following HBO exposure. The results suggest that the higher BGL following HBO exposure especially at SOD2 HET mice is in part due to reduction in GLUT1 as a consequence of lower HIF-1a expression. This may add part to the puzzle of the understanding the mechanism leading to CNS-OT.
Collapse
Affiliation(s)
- Ricarina Rabinovitz
- Israel Naval Medical Institute, Israel Defense Forces Medical Corps, Haifa, Israel
| | - Mirit Eynan
- Israel Naval Medical Institute, Israel Defense Forces Medical Corps, Haifa, Israel.
| |
Collapse
|
16
|
Averina OA, Kuznetsova SA, Permyakov OA, Sergiev PV. Animal Models of Mitochondrial Diseases Associated with Nuclear Gene Mutations. Acta Naturae 2023; 15:4-22. [PMID: 38234606 PMCID: PMC10790356 DOI: 10.32607/actanaturae.25442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 10/05/2023] [Indexed: 01/19/2024] Open
Abstract
Mitochondrial diseases (MDs) associated with nuclear gene mutations are part of a large group of inherited diseases caused by the suppression of energy metabolism. These diseases are of particular interest, because nuclear genes encode not only most of the structural proteins of the oxidative phosphorylation system (OXPHOS), but also all the proteins involved in the OXPHOS protein import from the cytoplasm and their assembly in mitochondria. Defects in any of these proteins can lead to functional impairment of the respiratory chain, including dysfunction of complex I that plays a central role in cellular respiration and oxidative phosphorylation, which is the most common cause of mitopathologies. Mitochondrial diseases are characterized by an early age of onset and a progressive course and affect primarily energy-consuming tissues and organs. The treatment of MDs should be initiated as soon as possible, but the diagnosis of mitopathologies is extremely difficult because of their heterogeneity and overlapping clinical features. The molecular pathogenesis of mitochondrial diseases is investigated using animal models: i.e. animals carrying mutations causing MD symptoms in humans. The use of mutant animal models opens new opportunities in the study of genes encoding mitochondrial proteins, as well as the molecular mechanisms of mitopathology development, which is necessary for improving diagnosis and developing approaches to drug therapy. In this review, we present the most recent information on mitochondrial diseases associated with nuclear gene mutations and animal models developed to investigate them.
Collapse
Affiliation(s)
- O. A. Averina
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - S. A. Kuznetsova
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - O. A. Permyakov
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| | - P. V. Sergiev
- Institute of Functional Genomics, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
- Department of Chemistry, Lomonosov Moscow State University, Moscow, 119991 Russian Federation
| |
Collapse
|
17
|
Riegger J, Schoppa A, Ruths L, Haffner-Luntzer M, Ignatius A. Oxidative stress as a key modulator of cell fate decision in osteoarthritis and osteoporosis: a narrative review. Cell Mol Biol Lett 2023; 28:76. [PMID: 37777764 PMCID: PMC10541721 DOI: 10.1186/s11658-023-00489-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 09/11/2023] [Indexed: 10/02/2023] Open
Abstract
During aging and after traumatic injuries, cartilage and bone cells are exposed to various pathophysiologic mediators, including reactive oxygen species (ROS), damage-associated molecular patterns, and proinflammatory cytokines. This detrimental environment triggers cellular stress and subsequent dysfunction, which not only contributes to the development of associated diseases, that is, osteoporosis and osteoarthritis, but also impairs regenerative processes. To counter ROS-mediated stress and reduce the overall tissue damage, cells possess diverse defense mechanisms. However, cellular antioxidative capacities are limited and thus ROS accumulation can lead to aberrant cell fate decisions, which have adverse effects on cartilage and bone homeostasis. In this narrative review, we address oxidative stress as a major driver of pathophysiologic processes in cartilage and bone, including senescence, misdirected differentiation, cell death, mitochondrial dysfunction, and impaired mitophagy by illustrating the consequences on tissue homeostasis and regeneration. Moreover, we elaborate cellular defense mechanisms, with a particular focus on oxidative stress response and mitophagy, and briefly discuss respective therapeutic strategies to improve cell and tissue protection.
Collapse
Affiliation(s)
- Jana Riegger
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany.
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Leonie Ruths
- Division for Biochemistry of Joint and Connective Tissue Diseases, Department of Orthopedics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, Ulm University Medical Center, 89081, Ulm, Germany
| |
Collapse
|
18
|
Maurya CK, Tapadia MG. Expanded polyQ aggregates interact with sarco-endoplasmic reticulum calcium ATPase and Drosophila inhibitor of apoptosis protein1 to regulate polyQ mediated neurodegeneration in Drosophila. Mol Cell Neurosci 2023; 126:103886. [PMID: 37567489 DOI: 10.1016/j.mcn.2023.103886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/04/2023] [Accepted: 08/08/2023] [Indexed: 08/13/2023] Open
Abstract
Polyglutamine (polyQ) induced neurodegeneration is one of the leading causes of progressive neurodegenerative disorders characterized clinically by deteriorating movement defects, psychiatric disability, and dementia. Calcium [Ca2+] homeostasis, which is essential for the functioning of neuronal cells, is disrupted under these pathological conditions. In this paper, we simulated Huntington's disease phenotype in the neuronal cells of the Drosophila eye and identified [Ca2+] pump, sarco-endoplasmic reticulum calcium ATPase (SERCA), as one of the genetic modifiers of the neurodegenerative phenotype. This paper shows genetic and molecular interaction between polyglutamine (polyQ) aggregates, SERCA and DIAP1. We present evidence that polyQ aggregates interact with SERCA and alter its dynamics, resulting in a decrease in cytosolic [Ca2+] and an increase in ER [Ca2+], and thus toxicity. Downregulating SERCA lowers the enhanced calcium levels in the ER and rescues, morphological and functional defects caused due to expanded polyQ repeats. Cell proliferation markers such as Yorkie (Yki), Scalloped (Sd), and phosphatidylinositol 3 kinases/protein kinase B (PI3K/Akt), also respond to varying levels of calcium due to genetic manipulations, adding to the amelioration of degeneration. These results imply that neurodegeneration due to expanded polyQ repeats is sensitive to SERCA activity, and its manipulation can be an important step toward its therapeutic measures.
Collapse
Affiliation(s)
- Chandan Kumar Maurya
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| | - Madhu G Tapadia
- Cytogenetics Laboratory, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
19
|
Tsesmelis K, Maity‐Kumar G, Croner D, Sprissler J, Tsesmelis M, Hein T, Baumann B, Wirth T. Accelerated aging in mice with astrocytic redox imbalance as a consequence of SOD2 deletion. Aging Cell 2023; 22:e13911. [PMID: 37609868 PMCID: PMC10497807 DOI: 10.1111/acel.13911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/08/2023] [Accepted: 05/31/2023] [Indexed: 08/24/2023] Open
Abstract
Aging of the central nervous system (CNS) leads to motoric and cognitive decline and increases the probability for neurodegenerative disease development. Astrocytes fulfill central homeostatic functions in the CNS including regulation of immune responses and metabolic support of neurons and oligodendrocytes. In this study, we investigated the effect of redox imbalance in astrocytes by using a conditional astrocyte-specific SOD2-deficient mouse model (SOD2ako ) and analyzed these animals at different stages of their life. SOD2ako mice did not exhibit any overt phenotype within the first postnatal weeks. However, already as young adults, they displayed progressive motoric impairments. Moreover, as these mice grew older, they exhibited signs of a progeroid phenotype and early death. Histological analysis in moribund SOD2ako mice revealed the presence of age-related brain alterations, neuroinflammation, neuronal damage and myelin impairment in brain and spinal cord. Additionally, transcriptome analysis of primary astrocytes revealed that SOD2 deletion triggered a hypometabolic state and promoted polarization toward A1-neurotoxic status, possibly underlying the neuronal and myelin deficits. Conclusively, our study identifies maintenance of ROS homeostasis in astrocytes as a critical prerequisite for physiological CNS aging.
Collapse
Affiliation(s)
| | - Gandhari Maity‐Kumar
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
- Institute for Diabetes and ObesityHelmholtz Diabetes Center at Helmholtz Zentrum MünchenNeuherbergGermany
| | - Dana Croner
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Jasmin Sprissler
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
- Department of Pediatrics and Adolescent MedicineUlm University Medical CenterUlmGermany
| | | | - Tabea Hein
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Bernd Baumann
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| | - Thomas Wirth
- Institute of Physiological ChemistryUniversity of UlmUlmGermany
| |
Collapse
|
20
|
Kuntic M, Kuntic I, Hahad O, Lelieveld J, Münzel T, Daiber A. Impact of air pollution on cardiovascular aging. Mech Ageing Dev 2023; 214:111857. [PMID: 37611809 DOI: 10.1016/j.mad.2023.111857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/19/2023] [Indexed: 08/25/2023]
Abstract
The world population is aging rapidly, and by some estimates, the number of people older than 60 will double in the next 30 years. With the increase in life expectancy, adverse effects of environmental exposures start playing a more prominent role in human health. Air pollution is now widely considered the most detrimental of all environmental risk factors, with some studies estimating that almost 20% of all deaths globally could be attributed to poor air quality. Cardiovascular diseases are the leading cause of death worldwide and will continue to account for the most significant percentage of non-communicable disease burden. Cardiovascular aging with defined pathomechanisms is a major trigger of cardiovascular disease in old age. Effects of environmental risk factors on cardiovascular aging should be considered in order to increase the health span and reduce the burden of cardiovascular disease in older populations. In this review, we explore the effects of air pollution on cardiovascular aging, from the molecular mechanisms to cardiovascular manifestations of aging and, finally, the age-related cardiovascular outcomes. We also explore the distinction between the effects of air pollution on healthy aging and disease progression. Future efforts should focus on extending the health span rather than the lifespan.
Collapse
Affiliation(s)
- Marin Kuntic
- University Medical Center Mainz, Department for Cardiology 1, Molecular Cardiology, Mainz, Germany
| | - Ivana Kuntic
- University Medical Center Mainz, Department for Cardiology 1, Molecular Cardiology, Mainz, Germany
| | - Omar Hahad
- University Medical Center Mainz, Department for Cardiology 1, Molecular Cardiology, Mainz, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany
| | - Jos Lelieveld
- Max Planck Institute for Chemistry, Atmospheric Chemistry, Mainz, Germany
| | - Thomas Münzel
- University Medical Center Mainz, Department for Cardiology 1, Molecular Cardiology, Mainz, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany.
| | - Andreas Daiber
- University Medical Center Mainz, Department for Cardiology 1, Molecular Cardiology, Mainz, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Mainz, Germany.
| |
Collapse
|
21
|
Soo SK, Rudich ZD, Ko B, Moldakozhayev A, AlOkda A, Van Raamsdonk JM. Biological resilience and aging: Activation of stress response pathways contributes to lifespan extension. Ageing Res Rev 2023; 88:101941. [PMID: 37127095 DOI: 10.1016/j.arr.2023.101941] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/06/2023] [Accepted: 04/28/2023] [Indexed: 05/03/2023]
Abstract
While aging was traditionally viewed as a stochastic process of damage accumulation, it is now clear that aging is strongly influenced by genetics. The identification and characterization of long-lived genetic mutants in model organisms has provided insights into the genetic pathways and molecular mechanisms involved in extending longevity. Long-lived genetic mutants exhibit activation of multiple stress response pathways leading to enhanced resistance to exogenous stressors. As a result, lifespan exhibits a significant, positive correlation with resistance to stress. Disruption of stress response pathways inhibits lifespan extension in multiple long-lived mutants representing different pathways of lifespan extension and can also reduce the lifespan of wild-type animals. Combined, this suggests that activation of stress response pathways is a key mechanism by which long-lived mutants achieve their extended longevity and that many of these pathways are also required for normal lifespan. These results highlight an important role for stress response pathways in determining the lifespan of an organism.
Collapse
Affiliation(s)
- Sonja K Soo
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Zenith D Rudich
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Bokang Ko
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Alibek Moldakozhayev
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Genetics, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, United States
| | - Abdelrahman AlOkda
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Jeremy M Van Raamsdonk
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada; Metabolic Disorders and Complications Program, and Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
22
|
Murphy E, Liu JC. Mitochondrial calcium and reactive oxygen species in cardiovascular disease. Cardiovasc Res 2023; 119:1105-1116. [PMID: 35986915 PMCID: PMC10411964 DOI: 10.1093/cvr/cvac134] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 05/26/2022] [Accepted: 06/02/2022] [Indexed: 08/11/2023] Open
Abstract
Cardiomyocytes are one of the most mitochondria-rich cell types in the body, with ∼30-40% of the cell volume being composed of mitochondria. Mitochondria are well established as the primary site of adenosine triphosphate (ATP) generation in a beating cardiomyocyte, generating up to 90% of its ATP. Mitochondria have many functions in the cell, which could contribute to susceptibility to and development of cardiovascular disease (CVD). Mitochondria are key players in cell metabolism, ATP production, reactive oxygen species (ROS) production, and cell death. Mitochondrial calcium (Ca2+) plays a critical role in many of these pathways, and thus the dynamics of mitochondrial Ca2+ are important in regulating mitochondrial processes. Alterations in these varied and in many cases interrelated functions play an important role in CVD. This review will focus on the interrelationship of mitochondrial energetics, Ca2+, and ROS and their roles in CVD. Recent insights into the regulation and dysregulation of these pathways have led to some novel therapeutic approaches.
Collapse
Affiliation(s)
- Elizabeth Murphy
- NHLBI, NIH, Bethesda, MD and Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| | - Julia C Liu
- NHLBI, NIH, Bethesda, MD and Department of Integrative Biology and Physiology, University of Minnesota, 2231 6th St. SE, Minneapolis, MN 55455, USA
| |
Collapse
|
23
|
Wörn M, Lämmer R, Lucio M, Michalke B, Rühl E, Hohberger B. The influence of trace elements on the therapeutic success of suprachoroidal draining devices. J Trace Elem Med Biol 2023; 78:127195. [PMID: 37207415 DOI: 10.1016/j.jtemb.2023.127195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 05/21/2023]
Abstract
PURPOSE The therapeutic success of minimal invasive glaucoma surgery (MIGS) is challenging due to many factors including fibrotic or occlusive events. Recent clinical data show sudden peaks of intraocular pressure (IOP) in the postoperative care of glaucoma patients after suprachoroidal draining stents. Yet, the reasons for the IOP peaks are speculative. As a link between trace elements and fibrosis had been previously observed in systemic disorders, the present study aimed to investigate the impact of trace elements on the therapeutic success of the suprachoroidal draining stents in patients with open-angle glaucoma (OAG). MATERIAL AND METHODS An analysis of a prospective single-center study was done: fifty-five eyes of patients with OAG (29 female, 26 male) underwent Cypass Micro-Stent implantation either as a stand-alone procedure or combined with cataract surgery. All patients underwent pre-operatively an ophthalmological examination which included slit lamp biomicroscopy and fundoscopy. IOP was measured by Goldmann applanation tonometry. Functional and morphometric data were assessed by Octopus G1-perimetry, which included measurement of retinal nerve fiber layer thickness (Spectralis OCT). Data of the patients' follow-ups were recorded during 18 months post-operatively. The therapeutic success of CyPass Micro-Stent was classified as 'success' (IOP reduction ≥20% compared to a pre-operative baseline without any medication), 'qualified success' (IOP reduction ≥20 % with same or lower additional eye medication), and 'failure' (IOP reduction ≤20 % or additional surgical treatment necessary). Aqueous humour was extracted once during surgery for analysis of the level of 14 trace elements: Copper (Cu), Cadmium (Cd), Cobalt (Co), Chromium (Cr), Iron (Fe), Lithium (Li), Magnesium (Mg), Manganese (Mn), Phosphorus (P), Lead (Pb), Titanium (Ti), Uranium (U), Vanadium (V), and Zinc (Zn). Analysis of the trace elements was done using an ELEMENT 2, ICP-sf-MS instrument (Thermo-Fisher Scientific, Bremen, Germany). Analysis of levels of trace elements was done across the patients' groups of the three subclasses of therapeutic success. Statistical investigations for substantial differences were conducted using the method of least squares to fit general linear models and mixed models. The last one for the repeated measurements of IOP. RESULTS Levels of Mg were significantly lower one month postoperatively in the success group (LS-Mean 1.30 mg/L) compared to the qualified success group (LS-Mean 1.22 mg/L; p-value = 0.04). Fe was significantly increased in the failure group (LS-Mean 2.07 µg/L) compared to the qualified success group (LS-Mean 1.64 µg/L; p-value = 0.019) after 3 months of follow-up. Additionally, Fe levels were significantly lower in the success group (LS-Mean 1.47 µg/L) compared to the failure cohort (LS-Mean 2.07 µg/L; p-value = 0.009). After a period of 18 months, significantly higher levels of Mn were observed in the success group (LS-Mean 1.24 µg/L) than in the failure group (LS Mean 0.30 µg/L, p-value = 0.019). CONCLUSION The present data might suggest that trace elements can influence therapeutic success of suprachoroidal draining devices postoperatively and thus offer first hints for potential novel therapeutic options.
Collapse
Affiliation(s)
- Maximilian Wörn
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Robert Lämmer
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Marianna Lucio
- BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Bernhard Michalke
- BioGeoChemistry, Helmholtz Zentrum München-German Research Center for Environmental Health, 85764 Neuherberg, Germany
| | - Eva Rühl
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Bettina Hohberger
- Department of Ophthalmology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
24
|
Bhaskaran S, Kumar G, Thadathil N, Piekarz KM, Mohammed S, Lopez SD, Qaisar R, Walton D, Brown JL, Murphy A, Smith N, Saunders D, Beckstead MJ, Plafker S, Lewis TL, Towner R, Deepa SS, Richardson A, Axtell RC, Van Remmen H. Neuronal deletion of MnSOD in mice leads to demyelination, inflammation and progressive paralysis that mimics phenotypes associated with progressive multiple sclerosis. Redox Biol 2023; 59:102550. [PMID: 36470129 PMCID: PMC9720104 DOI: 10.1016/j.redox.2022.102550] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022] Open
Abstract
Neuronal oxidative stress has been implicated in aging and neurodegenerative disease. Here we investigated the impact of elevated oxidative stress induced in mouse spinal cord by deletion of Mn-Superoxide dismutase (MnSOD) using a neuron specific Cre recombinase in Sod2 floxed mice (i-mn-Sod2 KO). Sod2 deletion in spinal cord neurons was associated with mitochondrial alterations and peroxide generation. Phenotypically, i-mn-Sod2 KO mice experienced hindlimb paralysis and clasping behavior associated with extensive demyelination and reduced nerve conduction velocity, axonal degeneration, enhanced blood brain barrier permeability, elevated inflammatory cytokines, microglia activation, infiltration of neutrophils and necroptosis in spinal cord. In contrast, spinal cord motor neuron number, innervation of neuromuscular junctions, muscle mass, and contractile function were not altered. Overall, our findings show that loss of MnSOD in spinal cord promotes a phenotype of demyelination, inflammation and progressive paralysis that mimics phenotypes associated with progressive multiple sclerosis.
Collapse
Affiliation(s)
- Shylesh Bhaskaran
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Gaurav Kumar
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, OK, USA
| | - Nidheesh Thadathil
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, OK, USA
| | - Katarzyna M Piekarz
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Sabira Mohammed
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Rizwan Qaisar
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Dorothy Walton
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Jacob L Brown
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Ashley Murphy
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Nataliya Smith
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, OK, USA
| | - Debra Saunders
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, OK, USA
| | - Michael J Beckstead
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Scott Plafker
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Tommy L Lewis
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA
| | - Rheal Towner
- Advanced Magnetic Resonance Center, Oklahoma Medical Research Foundation, OK, USA
| | - Sathyaseelan S Deepa
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Arlan Richardson
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA
| | - Robert C Axtell
- Arthritis & Clinical Immunology, Oklahoma Medical Research Foundation, OK, USA.
| | - Holly Van Remmen
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, OK, USA; Oklahoma City VA Medical Center, Oklahoma City, OK, USA.
| |
Collapse
|
25
|
Jain K, Gu SX, Hwa J. Cross talk on "endogenous SOD2 (superoxide dismutase) regulates platelet-dependent thrombin generation and thrombosis during aging" SOD2 in platelets: with age comes responsibility. JOURNAL OF THROMBOSIS AND HAEMOSTASIS : JTH 2023; 21:1077-1081. [PMID: 36716965 DOI: 10.1016/j.jtha.2023.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/29/2023]
Affiliation(s)
- Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, Connecticut, USA; Department of Laboratory Medicine, Yale University School of Medicine, Yale New Haven Hospital, New Haven, Connecticut, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
26
|
El Kodsi DN, Tokarew JM, Sengupta R, Lengacher NA, Chatterji A, Nguyen AP, Boston H, Jiang Q, Palmberg C, Pileggi C, Holterman CE, Shutinoski B, Li J, Fehr TK, LaVoie MJ, Ratan RR, Shaw GS, Takanashi M, Hattori N, Kennedy CR, Harper ME, Holmgren A, Tomlinson JJ, Schlossmacher MG. Parkin coregulates glutathione metabolism in adult mammalian brain. Acta Neuropathol Commun 2023; 11:19. [PMID: 36691076 PMCID: PMC9869535 DOI: 10.1186/s40478-022-01488-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 01/24/2023] Open
Abstract
We recently discovered that the expression of PRKN, a young-onset Parkinson disease-linked gene, confers redox homeostasis. To further examine the protective effects of parkin in an oxidative stress model, we first combined the loss of prkn with Sod2 haploinsufficiency in mice. Although adult prkn-/-//Sod2± animals did not develop dopamine cell loss in the S. nigra, they had more reactive oxidative species and a higher concentration of carbonylated proteins in the brain; bi-genic mice also showed a trend for more nitrotyrosinated proteins. Because these redox changes were seen in the cytosol rather than mitochondria, we next explored the thiol network in the context of PRKN expression. We detected a parkin deficiency-associated increase in the ratio of reduced glutathione (GSH) to oxidized glutathione (GSSG) in murine brain, PRKN-linked human cortex and several cell models. This shift resulted from enhanced recycling of GSSG back to GSH via upregulated glutathione reductase activity; it also correlated with altered activities of redox-sensitive enzymes in mitochondria isolated from mouse brain (e.g., aconitase-2; creatine kinase). Intriguingly, human parkin itself showed glutathione-recycling activity in vitro and in cells: For each GSSG dipeptide encountered, parkin regenerated one GSH molecule and was S-glutathionylated by the other (GSSG + P-SH [Formula: see text] GSH + P-S-SG), including at cysteines 59, 95 and 377. Moreover, parkin's S-glutathionylation was reversible by glutaredoxin activity. In summary, we found that PRKN gene expression contributes to the network of available thiols in the cell, including by parkin's participation in glutathione recycling, which involves a reversible, posttranslational modification at select cysteines. Further, parkin's impact on redox homeostasis in the cytosol can affect enzyme activities elsewhere, such as in mitochondria. We posit that antioxidant functions of parkin may explain many of its previously described, protective effects in vertebrates and invertebrates that are unrelated to E3 ligase activity.
Collapse
Affiliation(s)
- Daniel N El Kodsi
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jacqueline M Tokarew
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Rajib Sengupta
- Department of Biochemistry, Karolinska Institute, Stockholm, Sweden
- Amity Institute of Biotechnology, Amity University, Kolkata, West Bengal, India
| | - Nathalie A Lengacher
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Ajanta Chatterji
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Angela P Nguyen
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Snyder Institute, University of Calgary, Calgary, AB, Canada
| | - Heather Boston
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Qiubo Jiang
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Carina Palmberg
- Department of Biochemistry, Karolinska Institute, Stockholm, Sweden
| | - Chantal Pileggi
- Department of Biochemistry, Microbiology and Immunology Faculty of Medicine, and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Chet E Holterman
- Kidney Research Center, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Bojan Shutinoski
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Juan Li
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Travis K Fehr
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Matthew J LaVoie
- Department of Neurology, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Rajiv R Ratan
- Burke Neurological Institute, Weill Cornell Medical School, White Plains, NY, USA
| | - Gary S Shaw
- Department of Biochemistry, University of Western Ontario, London, ON, Canada
| | - Masashi Takanashi
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University School of Medicine, Tokyo, Japan
| | | | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology Faculty of Medicine, and Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, ON, Canada
| | - Arne Holmgren
- Department of Biochemistry, Karolinska Institute, Stockholm, Sweden
| | - Julianna J Tomlinson
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| | - Michael G Schlossmacher
- Program in Neuroscience, Ottawa Hospital Research Institute, Ottawa, ON, Canada.
- Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
- University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
- Division of Neurology, Department of Medicine, The Ottawa Hospital, Ottawa, ON, Canada.
| |
Collapse
|
27
|
Teixeira RB, Pfeiffer M, Zhang P, Shafique E, Rayta B, Karbasiafshar C, Ahsan N, Sellke FW, Abid MR. Reduction in mitochondrial ROS improves oxidative phosphorylation and provides resilience to coronary endothelium in non-reperfused myocardial infarction. Basic Res Cardiol 2023; 118:3. [PMID: 36639609 PMCID: PMC9839395 DOI: 10.1007/s00395-022-00976-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 12/30/2022] [Accepted: 12/30/2022] [Indexed: 01/15/2023]
Abstract
Recent studies demonstrated that mitochondrial antioxidant MnSOD that reduces mitochondrial (mito) reactive oxygen species (ROS) helps maintain an optimal balance between sub-cellular ROS levels in coronary vascular endothelial cells (ECs). However, it is not known whether EC-specific mito-ROS modulation provides resilience to coronary ECs after a non-reperfused acute myocardial infarction (MI). This study examined whether a reduction in endothelium-specific mito-ROS improves the survival and proliferation of coronary ECs in vivo. We generated a novel conditional binary transgenic animal model that overexpresses (OE) mitochondrial antioxidant MnSOD in an EC-specific manner (MnSOD-OE). EC-specific MnSOD-OE was validated in heart sections and mouse heart ECs (MHECs). Mitosox and mito-roGFP assays demonstrated that MnSOD-OE resulted in a 50% reduction in mito-ROS in MHEC. Control and MnSOD-OE mice were subject to non-reperfusion MI surgery, echocardiography, and heart harvest. In post-MI hearts, MnSOD-OE promoted EC proliferation (by 2.4 ± 0.9 fold) and coronary angiogenesis (by 3.4 ± 0.9 fold), reduced myocardial infarct size (by 27%), and improved left ventricle ejection fraction (by 16%) and fractional shortening (by 20%). Interestingly, proteomic and Western blot analyses demonstrated upregulation in mitochondrial complex I and oxidative phosphorylation (OXPHOS) proteins in MnSOD-OE MHECs. These MHECs also showed increased mitochondrial oxygen consumption rate (OCR) and membrane potential. These findings suggest that mito-ROS reduction in EC improves coronary angiogenesis and cardiac function in non-reperfused MI, which are associated with increased activation of OXPHOS in EC-mitochondria. Activation of an energy-efficient mechanism in EC may be a novel mechanism to confer resilience to coronary EC during MI.
Collapse
Affiliation(s)
- Rayane Brinck Teixeira
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA
| | - Melissa Pfeiffer
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA
| | - Peng Zhang
- Vascular Research Laboratory/Providence VA Medical Center and Department of Medicine, Alpert Medical School of Brown University, Providence, RI, USA
| | - Ehtesham Shafique
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA
| | - Bonnie Rayta
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA
| | - Catherine Karbasiafshar
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA
| | - Nagib Ahsan
- Division of Biology and Medicine, Alpert Medical School, Brown University, Providence, RI, 02903, USA
- Proteomics Core Facility, Center for Cancer Research and Development, Rhode Island Hospital, Providence, RI, 02903, USA
- Department of Chemistry and Biochemistry, Mass Spectrometry, Proteomics and Metabolomics Core Facility, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, OK, USA
| | - Frank W Sellke
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Warren Alpert Medical School, 1 Hoppin Street, Providence, RI, 02903, USA.
| |
Collapse
|
28
|
Booth LK, Redgrave RE, Tual-Chalot S, Spyridopoulos I, Phillips HM, Richardson GD. Heart Disease and Ageing: The Roles of Senescence, Mitochondria, and Telomerase in Cardiovascular Disease. Subcell Biochem 2023; 103:45-78. [PMID: 37120464 DOI: 10.1007/978-3-031-26576-1_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/01/2023]
Abstract
During ageing molecular damage leads to the accumulation of several hallmarks of ageing including mitochondrial dysfunction, cellular senescence, genetic instability and chronic inflammation, which contribute to the development and progression of ageing-associated diseases including cardiovascular disease. Consequently, understanding how these hallmarks of biological ageing interact with the cardiovascular system and each other is fundamental to the pursuit of improving cardiovascular health globally. This review provides an overview of our current understanding of how candidate hallmarks contribute to cardiovascular diseases such as atherosclerosis, coronary artery disease and subsequent myocardial infarction, and age-related heart failure. Further, we consider the evidence that, even in the absence of chronological age, acute cellular stress leading to accelerated biological ageing expedites cardiovascular dysfunction and impacts on cardiovascular health. Finally, we consider the opportunities that modulating hallmarks of ageing offer for the development of novel cardiovascular therapeutics.
Collapse
Affiliation(s)
- Laura K Booth
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Rachael E Redgrave
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Ioakim Spyridopoulos
- Translational and Clinical Research Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Helen M Phillips
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK
| | - Gavin D Richardson
- Biosciences Institute, Vascular Biology and Medicine Theme, Newcastle University, Newcastle upon Tyne, UK.
| |
Collapse
|
29
|
Debeljak M, Riel S, Lin MT, Eshleman JR, Paller CJ. Analytical Validation of SOD2 Genotyping. Methods Protoc 2022; 6:mps6010004. [PMID: 36648953 PMCID: PMC9844328 DOI: 10.3390/mps6010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 01/03/2023] Open
Abstract
Manganese superoxide dismutase-2 (SOD2) plays a crucial role in cells' protection against mitochondrial oxidative damage. A genetic polymorphism in the mitochondrial targeting sequence of the SOD2 gene has been implicated in various diseases, including prostate cancer. Paller et al. have shown an increase in prostate-specific antigen (PSA) doubling time in patients with the Ala/Ala (wildtype) genotype when treated with pomegranate/grape extract antioxidants. We developed and validated a pyrosequencing assay that detects the common germline SOD2 SNP (rs_4880) with the aim of identifying men with castrate-resistant prostate cancer eligible for an antioxidant therapy clinical trial. We first selected 37 samples from the 1000 genomes study with known genotypes determined using Illumina-based sequencing and confirmed them by Sanger sequencing. In a blinded design, we then performed the new pyrosequencing assay on these samples and assigned genotypes. Genotypes for all 37 samples (13 homozygous Ala, 12 heterozygous Ala/Val, and 12 homozygous Val) were all concordant by pyrosequencing. The pyrosequencing assay has been live since May 2018 and has proven to be robust and accurate.
Collapse
Affiliation(s)
- Marija Debeljak
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Stacy Riel
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ming-Tseh Lin
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - James R. Eshleman
- Department of Pathology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Correspondence: (J.R.E.); (C.J.P.)
| | - Channing J. Paller
- Department of Oncology, The Sol Goldman Pancreatic Cancer Research Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
- Correspondence: (J.R.E.); (C.J.P.)
| |
Collapse
|
30
|
Lee G, Banik A, Eum J, Hwang BJ, Kwon SH, Kee Y. Ipconazole Disrupts Mitochondrial Homeostasis and Alters GABAergic Neuronal Development in Zebrafish. Int J Mol Sci 2022; 24:496. [PMID: 36613936 PMCID: PMC9820214 DOI: 10.3390/ijms24010496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/15/2022] [Accepted: 12/23/2022] [Indexed: 12/29/2022] Open
Abstract
Ipconazole, a demethylation inhibitor of fungal ergosterol biosynthesis, is widely used in modern agriculture for foliar and seed treatment, and is authorized for use in livestock feed. Waste from ipconazole treatment enters rivers and groundwater through disposal and rain, posing potential toxicity to humans and other organisms. Its metabolites remain stable under standard hydrolysis conditions; however, their neurodevelopmental toxicity is unknown. We investigated the potential neurodevelopmental toxicity of ipconazole pesticides in zebrafish (Danio rerio). Our behavioral monitoring demonstrated that the locomotive activity of ipconazole-exposed zebrafish larvae was reduced during early development, even when morphological abnormalities were undetected. Molecular profiling demonstrated that the mitochondrial-specific antioxidants, superoxide dismutases 1 and 2, and the genes essential for mitochondrial genome maintenance and functions were specifically reduced in ipconazole-treated (0.02 μg/mL) embryos, suggesting underlying ipconazole-driven oxidative stress. Consistently, ipconazole treatment substantially reduced hsp70 expression and increased ERK1/2 phosphorylation in a dose-dependent manner. Interrupted gad1b expression confirmed that GABAergic inhibitory neurons were dysregulated at 0.02 μg/mL ipconazole, whereas glutamatergic excitatory and dopaminergic neurons remained unaffected, resulting in an uncoordinated neural network. Additionally, ipconazole-treated (2 μg/mL) embryos exhibited caspase-independent cell death. This suggests that ipconazole has the potential to alter neurodevelopment by dysregulating mitochondrial homeostasis.
Collapse
Affiliation(s)
- Giyoung Lee
- Department of Biomedical Science, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Amit Banik
- Interdisciplinary Graduate Program in Environmental and Biomedical Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Juneyong Eum
- Interdisciplinary Graduate Program in Environmental and Biomedical Convergence, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Byung Joon Hwang
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| | - Seung-Hae Kwon
- Korea Basic Science Institute Seoul Center, Seoul 02841, Republic of Korea
| | - Yun Kee
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon 24341, Republic of Korea
| |
Collapse
|
31
|
Liu M, Sun X, Chen B, Dai R, Xi Z, Xu H. Insights into Manganese Superoxide Dismutase and Human Diseases. Int J Mol Sci 2022; 23:ijms232415893. [PMID: 36555531 PMCID: PMC9786916 DOI: 10.3390/ijms232415893] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/09/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox equilibria and the modulation of redox signalling play crucial roles in physiological processes. Overproduction of reactive oxygen species (ROS) disrupts the body's antioxidant defence, compromising redox homeostasis and increasing oxidative stress, leading to the development of several diseases. Manganese superoxide dismutase (MnSOD) is a principal antioxidant enzyme that protects cells from oxidative damage by converting superoxide anion radicals to hydrogen peroxide and oxygen in mitochondria. Systematic studies have demonstrated that MnSOD plays an indispensable role in multiple diseases. This review focuses on preclinical evidence that describes the mechanisms of MnSOD in diseases accompanied with an imbalanced redox status, including fibrotic diseases, inflammation, diabetes, vascular diseases, neurodegenerative diseases, and cancer. The potential therapeutic effects of MnSOD activators and MnSOD mimetics are also discussed. Targeting this specific superoxide anion radical scavenger may be a clinically beneficial strategy, and understanding the therapeutic role of MnSOD may provide a positive insight into preventing and treating related diseases.
Collapse
Affiliation(s)
- Mengfan Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Xueyang Sun
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Boya Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Rongchen Dai
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
| | - Zhichao Xi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
- Correspondence: (Z.X.); (H.X.)
| | - Hongxi Xu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Engineering Research Center, Shanghai Colleges for TCM New Drug Discovery, Shanghai 201203, China
- Correspondence: (Z.X.); (H.X.)
| |
Collapse
|
32
|
Prieux-Klotz C, Chédotal H, Zoumpoulaki M, Chouzenoux S, Chêne C, Lopez-Sanchez A, Thomas M, Ranjan Sahoo P, Policar C, Batteux F, Bertrand HC, Nicco C, Coriat R. A New Manganese Superoxide Dismutase Mimetic Improves Oxaliplatin-Induced Neuropathy and Global Tolerance in Mice. Int J Mol Sci 2022; 23:12938. [PMID: 36361753 PMCID: PMC9658974 DOI: 10.3390/ijms232112938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 09/08/2024] Open
Abstract
Reactive oxygen species (ROS) are produced by every aerobic cell during mitochondrial oxidative metabolism as well as in cellular response to xenobiotics, cytokines, and bacterial invasion. Superoxide Dismutases (SOD) are antioxidant proteins that convert superoxide anions (O2•-) to hydrogen peroxide (H2O2) and dioxygen. Using the differential in the level of oxidative stress between normal and cancer cells, SOD mimetics can show an antitumoral effect and prevent oxaliplatin-induced peripheral neuropathy. New Pt(IV) conjugate prodrugs (OxPt-x-Mn1C1A (x = 1, 1-OH, 2)), combining oxaliplatin and a Mn SOD mimic (MnSODm Mn1C1A) with a covalent link, were designed. Their stability in buffer and in the presence of sodium ascorbate was studied. In vitro, their antitumoral activity was assessed by the viability and ROS production of tumor cell lines (CT16, HCT 116, KC) and fibroblasts (primary culture and NIH 3T3). In vivo, a murine model of colorectal cancer was created with subcutaneous injection of CT26 cells in Balb/c mice. Tumor size and volume were measured weekly in four groups: vehicle, oxaliplatin, and oxaliplatin associated with MnSODm Mn1C1A and the bis-conjugate OxPt-2-Mn1C1A. Oxaliplatin-induced peripheral neuropathy (OIPN) was assessed using a Von Frey test reflecting chronic hypoalgesia. Tolerance to treatment was assessed with a clinical score including four items: weight loss, weariness, alopecia, and diarrhea. In vitro, Mn1C1A associated with oxaliplatin and Pt(IV) conjugates treatment induced significantly higher production of H2O2 in all cell lines and showed a significant improvement of the antitumoral efficacy compared to oxaliplatin alone. In vivo, the association of Mn1C1A to oxaliplatin did not decrease its antitumoral activity, while OxPt-2-Mn1C1A had lower antitumoral activity than oxaliplatin alone. Mn1C1A associated with oxaliplatin significantly decreased OIPN and also improved global clinical tolerance of oxaliplatin. A neuroprotective effect was observed, associated with a significantly improved tolerance to oxaliplatin without impairing its antitumoral activity.
Collapse
Affiliation(s)
- Caroline Prieux-Klotz
- Institut Cochin, INSERM U 1016 CNRS UMR 8104, Université de Paris, 75005 Paris, France
- Percy Military Hospital, Gastroenterology, 101 Avenue Henri Barbusse, 92140 Clamart, France
- Laboratoire des Biomolécules, LBM, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Henri Chédotal
- Laboratoire des Biomolécules, LBM, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Martha Zoumpoulaki
- Laboratoire des Biomolécules, LBM, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Sandrine Chouzenoux
- Institut Cochin, INSERM U 1016 CNRS UMR 8104, Université de Paris, 75005 Paris, France
| | - Charlotte Chêne
- Institut Cochin, INSERM U 1016 CNRS UMR 8104, Université de Paris, 75005 Paris, France
| | - Alvaro Lopez-Sanchez
- Laboratoire des Biomolécules, LBM, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Marine Thomas
- Institut Cochin, INSERM U 1016 CNRS UMR 8104, Université de Paris, 75005 Paris, France
| | - Priya Ranjan Sahoo
- Laboratoire des Biomolécules, LBM, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Clotilde Policar
- Laboratoire des Biomolécules, LBM, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Frédéric Batteux
- Institut Cochin, INSERM U 1016 CNRS UMR 8104, Université de Paris, 75005 Paris, France
| | - Hélène C. Bertrand
- Laboratoire des Biomolécules, LBM, Département de Chimie, Ecole Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005 Paris, France
| | - Carole Nicco
- Institut Cochin, INSERM U 1016 CNRS UMR 8104, Université de Paris, 75005 Paris, France
| | - Romain Coriat
- Institut Cochin, INSERM U 1016 CNRS UMR 8104, Université de Paris, 75005 Paris, France
- Gastroenterology, Cochin Hospital AP-HP, Université de Paris, 75014 Paris, France
| |
Collapse
|
33
|
Qrareya AN, Wise NS, Hodges ER, Mahdi F, Stewart JA, Paris JJ. HIV-1 Tat Upregulates the Receptor for Advanced Glycation End Products and Superoxide Dismutase-2 in the Heart of Transgenic Mice. Viruses 2022; 14:v14102191. [PMID: 36298745 PMCID: PMC9607872 DOI: 10.3390/v14102191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/23/2022] [Accepted: 09/27/2022] [Indexed: 11/29/2022] Open
Abstract
Cardiovascular disorder (CVD) is a common comorbidity in people living with HIV (PLWH). Although the underlying mechanisms are unknown, virotoxic HIV proteins, such as the trans-activator of transcription (Tat), likely contribute to CVD pathogenesis. Tat expression in mouse myocardium has been found to induce cardiac dysfunction and increase markers of endothelial toxicity. However, the role that Tat may play in the development of CVD pathogenesis is unclear. The capacity for Tat to impact cardiac function was assessed using AC16 human cardiomyocyte cells and adult male and female transgenic mice that conditionally expressed Tat [Tat(+)], or did not [Tat(-)]. In AC16 cardiomyocytes, Tat increased intracellular calcium. In Tat(+) mice, Tat expression was detected in both atrial and ventricular heart tissue. Tat(+) mice demonstrated an increased expression of the receptor for advanced glycation end products and superoxide dismutase-2 (SOD-2) in ventricular tissues compared to Tat(-) controls. No changes in SOD-1 or α-smooth muscle actin were observed. Despite Tat-mediated changes at the cellular level, no changes in echocardiographic measures were detected. Tat(+) mice had a greater proportion of ventricular mast cells and collagen; however, doxycycline exposure offset the latter effect. These data suggest that Tat exposure promotes cellular changes that can precede progression to CVD.
Collapse
Affiliation(s)
- Alaa N. Qrareya
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Nason S. Wise
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Emmanuel R. Hodges
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Fakhri Mahdi
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - James A. Stewart
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Research Institute of Pharmaceutical Sciences, University of Mississippi, Oxford, MS 38677, USA
- Correspondence: (J.A.S.J.); (J.J.P.); Tel.: +1-662-915-2309 (J.A.S.J.); +1-662-915-3096 (J.J.P.)
| | - Jason J. Paris
- Department of BioMolecular Sciences, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
- Research Institute of Pharmaceutical Sciences, University of Mississippi, Oxford, MS 38677, USA
- Correspondence: (J.A.S.J.); (J.J.P.); Tel.: +1-662-915-2309 (J.A.S.J.); +1-662-915-3096 (J.J.P.)
| |
Collapse
|
34
|
Fernández-Espejo E. Is there a halo-enzymopathy in Parkinson's disease? Neurologia 2022; 37:661-667. [PMID: 30952477 DOI: 10.1016/j.nrl.2018.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/12/2018] [Accepted: 12/20/2018] [Indexed: 11/18/2022] Open
Abstract
Laboratory studies identified changes in the metabolism of halogens in the serum and cerebrospinal fluid (CSF) of patients with Parkinson's disease, which indicates the presence of «accelerated self-halogenation» of CSF and/or an increase in haloperoxidases, specifically serum thyroperoxidase and CSF lactoperoxidase. Furthermore, an excess of some halogenated derivatives, such as advanced oxygenation protein products (AOPP), has been detected in the CSF and serum. «Accelerated self-halogenation» and increased levels of haloperoxidases and AOPP proteins indicate that halogenative stress is present in Parkinson's disease. In addition, 3-iodo-L-tyrosine, a halogenated derivative, shows «parkinsonian» toxicity in experimental models, since it has been observed to induce α-synuclein aggregation and damage to dopaminergic neurons in the mouse brain and intestine. The hypothesis is that patients with Parkinson's disease display halogenative stress related to a haloenzymatic alteration of the synthesis or degradation of oxyacid of halogens and their halogenated derivatives. This halogenative stress would be related to nervous system damage.
Collapse
Affiliation(s)
- E Fernández-Espejo
- Laboratorio de Neurofisiología y Neurología Molecular, Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla, Sevilla, España.
| |
Collapse
|
35
|
Niveditha S, Shivanandappa T. Potentiation of paraquat toxicity by inhibition of the antioxidant defenses and protective effect of the natural antioxidant, 4-hydroxyisopthalic acid in Drosophila melanogaster. Comp Biochem Physiol C Toxicol Pharmacol 2022; 259:109399. [PMID: 35753646 DOI: 10.1016/j.cbpc.2022.109399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/23/2022] [Accepted: 06/19/2022] [Indexed: 11/25/2022]
Abstract
Exposure to pesticides such as paraquat (PQ) is known to induce oxidative stress-mediated damage, which is implicated in neurodegenerative diseases. The antioxidant enzymes are part of the endogenous defense mechanisms capable of protecting against oxidative damage, and down-regulation of these enzymes results in elevated oxidative stress. In this study, we have evaluated the protective action of 4-hydroxyisophthalic acid (DHA-I), a novel bioactive molecule from the roots of D. hamiltonii, against PQ toxicity and demonstrated the protective role of endogenous antioxidant enzymes under the condition of oxidative stress using Drosophila model. The activity of the major antioxidant enzymes, superoxide dismutase 1 (SOD1) and catalase, was suppressed either by RNAi-mediated post transcriptional gene silencing or chemical inhibition. With the decreased in vivo activity of either SOD1 or catalase, Drosophila exhibited hypersensitivity to PQ toxicity, demonstrating the essential role of antioxidant enzymes in the mechanism of defense against PQ-induced oxidative stress. Dietary supplementation of DHA-I increased the resistance of Drosophila depleted in either SOD1 or catalase to PQ toxicity. Enhanced survival of flies against PQ toxicity indicates the protective role of DHA-I against oxidative stress-mediated damage under the condition of compromised antioxidant defenses.
Collapse
Affiliation(s)
- S Niveditha
- Neurobiology laboratory, Department of Zoology, University of Mysore, Manasagangotri, Mysuru 570006, Karnataka, India
| | - T Shivanandappa
- Neurobiology laboratory, Department of Zoology, University of Mysore, Manasagangotri, Mysuru 570006, Karnataka, India.
| |
Collapse
|
36
|
Wang H, Montague HR, Hess HN, Zhang Y, Aguilar GL, Dunham RA, Butts IAE, Wang X. Transcriptome Analysis Reveals Key Gene Expression Changes in Blue Catfish Sperm in Response to Cryopreservation. Int J Mol Sci 2022; 23:ijms23147618. [PMID: 35886966 PMCID: PMC9316979 DOI: 10.3390/ijms23147618] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
The hybrids of female channel catfish (Ictalurus punctatus) and male blue catfish (I. furcatus) account for >50% of US catfish production due to superior growth, feed conversion, and disease resistance compared to both parental species. However, these hybrids can rarely be naturally spawned. Sperm collection is a lethal procedure, and sperm samples are now cryopreserved for fertilization needs. Previous studies showed that variation in sperm quality causes variable embryo hatch rates, which is the limiting factor in hybrid catfish breeding. Biomarkers as indicators for sperm quality and reproductive success are currently lacking. To address this, we investigated expression changes caused by cryopreservation using transcriptome profiles of fresh and cryopreserved sperm. Sperm quality measurements revealed that cryopreservation significantly increased oxidative stress levels and DNA fragmentation, and reduced sperm kinematic parameters. The present RNA-seq study identified 849 upregulated genes after cryopreservation, including members of all five complexes in the mitochondrial electron transport chain, suggesting a boost in oxidative phosphorylation activities, which often lead to excessive production of reactive oxygen species (ROS) associated with cell death. Interestingly, functional enrichment analyses revealed compensatory changes in gene expression after cryopreservation to offset detrimental effects of ultra-cold storage: MnSOD was induced to control ROS production; chaperones and ubiquitin ligases were upregulated to correct misfolded proteins or direct them to degradation; negative regulators of apoptosis, amide biosynthesis, and cilium-related functions were also enriched. Our study provides insight into underlying molecular mechanisms of sperm cryoinjury and lays a foundation to further explore molecular biomarkers on cryo-survival and gamete quality.
Collapse
Affiliation(s)
- Haolong Wang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (H.W.); (Y.Z.)
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
| | - Helen R. Montague
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Hana N. Hess
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Ying Zhang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (H.W.); (Y.Z.)
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
| | - Gavin L. Aguilar
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Rex A. Dunham
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
| | - Ian A. E. Butts
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
- School of Fisheries, Aquaculture and Aquatic Sciences, Auburn University, Auburn, AL 36849, USA
- Correspondence: (I.A.E.B.); (X.W.); Tel.: +1-344-728-7745 (I.A.E.B.); +1-344-844-7511 (X.W.)
| | - Xu Wang
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA; (H.W.); (Y.Z.)
- Alabama Agricultural Experiment Station, Auburn, AL 36849, USA; (H.R.M.); (H.N.H.); (G.L.A.); (R.A.D.)
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
- Correspondence: (I.A.E.B.); (X.W.); Tel.: +1-344-728-7745 (I.A.E.B.); +1-344-844-7511 (X.W.)
| |
Collapse
|
37
|
Hispanic ethnicity and the rs4880 variant in SOD2 are associated with elevated liver enzymes and bilirubin levels in children receiving asparaginase-containing chemotherapy for acute lymphoblastic leukemia. Biomed Pharmacother 2022; 150:113000. [PMID: 35658244 PMCID: PMC9450009 DOI: 10.1016/j.biopha.2022.113000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/11/2022] [Accepted: 04/17/2022] [Indexed: 01/29/2023] Open
Abstract
Asparaginase is an integral component of acute lymphoblastic leukemia (ALL)3 treatment. Hepatotoxicity related to asparaginase is one of the most common treatment-related toxicities in ALL therapy. Hispanic children are at higher risk of developing ALL, and toxicities from ALL therapy. The rs4880 variant in the superoxide dismutase 2 (SOD2)4 gene, a critical mitochondrial enzyme that protects cells against oxidative stress, was found to be associated with increased incidence of asparaginase-related hepatotoxicity in adult cohort of largely White non-Hispanics patients with ALL. The risk genotype (rs4880-CC) is more frequent among adult Hispanic patients with ALL. To assess the prevalence of hepatotoxicity and risk genotype among pediatric patients with ALL, particularly of Hispanic ethnicity, we conducted a prospective study of 143 pediatric patients with ALL (62.2% Hispanic). Bilirubin and hepatic transaminase levels were collected at different times during multiagent therapy including asparaginase treatment. Germline DNA blood samples were genotyped for the SOD2 rs4880. We found that the frequency of hepatotoxicity and the rs4880-CC risk genotype are higher in Hispanic patients than non-Hispanic. Patients with the CC genotype exhibit higher bilirubin and hepatic transaminase levels compared with patients with the TT and CT genotypes. In a multivariate Cox analysis, Hispanic ethnicity was identified as a strong predictor of hepatotoxicity (hazard ratio [HR] = 1.9, 95% confidence interval [95% CI] 1.0-3.5, p = 0.05). Altogether, these findings demonstrate that hepatotoxicity is highly prevalent among Hispanic pediatric patients with ALL, and those with rs4880-CC genotype.
Collapse
|
38
|
Protective Effects of N-Acetylcysteine on Lipopolysaccharide-Induced Respiratory Inflammation and Oxidative Stress. Antioxidants (Basel) 2022; 11:antiox11050879. [PMID: 35624744 PMCID: PMC9137500 DOI: 10.3390/antiox11050879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/23/2022] [Accepted: 04/25/2022] [Indexed: 01/18/2023] Open
Abstract
As the leading cause of bovine respiratory disease (BRD), bacterial pneumonia can result in tremendous losses in the herd farming industry worldwide. N-acetylcysteine (NAC), an acetylated precursor of the amino acid L-cysteine, has been reported to have anti-inflammatory and antioxidant properties. To explore the protective effect and underlying mechanisms of NAC in ALI, we investigated its role in lipopolysaccharide (LPS)-induced bovine embryo tracheal cells (EBTr) and mouse lung injury models. We found that NAC pretreatment attenuated LPS-induced inflammation in EBTr and mouse models. Moreover, LPS suppressed the expression of oxidative-related factors in EBTr and promoted gene expression and the secretion of inflammatory cytokines. Conversely, the pretreatment of NAC alleviated the secretion of inflammatory cytokines and decreased their mRNA levels, maintaining stable levels of antioxidative gene expression. In vivo, NAC helped LPS-induced inflammatory responses and lung injury in ALI mice. The relative protein concentration, total cells, and percentage of neutrophils in BALF; the level of secretion of IL-6, IL-8, TNF-α, and IL-1β; MPO activity; lung injury score; and the expression level of inflammatory-related genes were decreased significantly in the NAC group compared with the LPS group. NAC also ameliorated LPS-induced mRNA level changes in antioxidative genes. In conclusion, our findings suggest that NAC affects the inflammatory and oxidative response, alleviating LPS-induced EBTr inflammation and mouse lung injury, which offers a natural therapeutic strategy for BRD.
Collapse
|
39
|
Superoxide Radicals in the Execution of Cell Death. Antioxidants (Basel) 2022; 11:antiox11030501. [PMID: 35326151 PMCID: PMC8944419 DOI: 10.3390/antiox11030501] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/01/2022] [Accepted: 03/01/2022] [Indexed: 12/24/2022] Open
Abstract
Superoxide is a primary oxygen radical that is produced when an oxygen molecule receives one electron. Superoxide dismutase (SOD) plays a primary role in the cellular defense against an oxidative insult by ROS. However, the resulting hydrogen peroxide is still reactive and, in the presence of free ferrous iron, may produce hydroxyl radicals and exacerbate diseases. Polyunsaturated fatty acids are the preferred target of hydroxyl radicals. Ferroptosis, a type of necrotic cell death induced by lipid peroxides in the presence of free iron, has attracted considerable interest because of its role in the pathogenesis of many diseases. Radical electrons, namely those released from mitochondrial electron transfer complexes, and those produced by enzymatic reactions, such as lipoxygenases, appear to cause lipid peroxidation. While GPX4 is the most potent anti-ferroptotic enzyme that is known to reduce lipid peroxides to alcohols, other antioxidative enzymes are also indirectly involved in protection against ferroptosis. Moreover, several low molecular weight compounds that include α-tocopherol, ascorbate, and nitric oxide also efficiently neutralize radical electrons, thereby suppressing ferroptosis. The removal of radical electrons in the early stages is of primary importance in protecting against ferroptosis and other diseases that are related to oxidative stress.
Collapse
|
40
|
Biswas K, Alexander K, Francis MM. Reactive Oxygen Species: Angels and Demons in the Life of a Neuron. NEUROSCI 2022; 3:130-145. [PMID: 39484669 PMCID: PMC11523706 DOI: 10.3390/neurosci3010011] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 03/13/2022] [Indexed: 11/03/2024] Open
Abstract
Reactive oxygen species (ROS) have emerged as regulators of key processes supporting neuronal growth, function, and plasticity across lifespan. At normal physiological levels, ROS perform important roles as secondary messengers in diverse molecular processes such as regulating neuronal differentiation, polarization, synapse maturation, and neurotransmission. In contrast, high levels of ROS are toxic and can ultimately lead to cell death. Excitable cells, such as neurons, often require high levels of metabolic activity to perform their functions. As a consequence, these cells are more likely to produce high levels of ROS, potentially enhancing their susceptibility to oxidative damage. In addition, because neurons are generally post-mitotic, they may be subject to accumulating oxidative damage. Thus, maintaining tight control over ROS concentration in the nervous system is essential for proper neuronal development and function. We are developing a more complete understanding of the cellular and molecular mechanisms for control of ROS in these processes. This review focuses on ROS regulation of the developmental and functional properties of neurons, highlighting recent in vivo studies. We also discuss the current evidence linking oxidative damage to pathological conditions associated with neurodevelopmental and neurodegenerative disorders.
Collapse
Affiliation(s)
- Kasturi Biswas
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (K.B.); (K.A.)
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Kellianne Alexander
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (K.B.); (K.A.)
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Michael M Francis
- Department of Neurobiology, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; (K.B.); (K.A.)
- Graduate Program in Neuroscience, Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
41
|
Androgen-Dependent Prostate Cancer Cells Reprogram Their Metabolic Signature upon GLUT1 Upregulation by Manganese Superoxide Dismutase. Antioxidants (Basel) 2022; 11:antiox11020313. [PMID: 35204196 PMCID: PMC8868133 DOI: 10.3390/antiox11020313] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/27/2022] [Accepted: 01/31/2022] [Indexed: 11/17/2022] Open
Abstract
Prostate cancer is the second leading cause of cancer in men across the globe. The prostate gland accounts for some unique glycolytic metabolic characteristics, which causes the metabolic features of prostate tumor initiation and progression to remain poorly characterized. The mitochondrial superoxide dismutase (SOD2) is one of the major redox metabolism regulators. This study points out SOD2 as one major regulator for both redox and glycolytic metabolism in prostate cancer. SOD2 overexpression increases glucose transporter GLUT-1 and glucose uptake. This is not an insulin-mediated effect and seems to be sex-dependent, being present in male mice only. This event concurs with a series of substantial metabolic rearrangements at cytoplasmic and mitochondrial level. A concomitant decrease in glycolytic and pentose phosphate activity, and an increase in electron transfer in the mitochondrial electronic chain, were observed. The Krebs Cycle is altered to produce amino-acid intermediates by decreasing succinate dehydrogenase. This in turn generates a 13-fold increase in the oncometabolite succinate. The protein energy sensor AMPK is decreased at basal and phosphorylated levels in response to glucose deprivation. Finally, preliminary results in prostate cancer patients indicate that glandular areas presenting high levels of SOD2 show a very strong correlation with GLUT-1 protein levels (R2 = 0.287 p-value < 0.0001), indicating that in patients there may exist an analogous phenomenon to those observed in cell culture and mice.
Collapse
|
42
|
Ayer A, Fazakerley DJ, James DE, Stocker R. The role of mitochondrial reactive oxygen species in insulin resistance. Free Radic Biol Med 2022; 179:339-362. [PMID: 34775001 DOI: 10.1016/j.freeradbiomed.2021.11.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/31/2021] [Accepted: 11/06/2021] [Indexed: 12/21/2022]
Abstract
Insulin resistance is one of the earliest pathological features of a suite of diseases including type 2 diabetes collectively referred to as metabolic syndrome. There is a growing body of evidence from both pre-clinical studies and human cohorts indicating that reactive oxygen species, such as the superoxide radical anion and hydrogen peroxide are key players in the development of insulin resistance. Here we review the evidence linking mitochondrial reactive oxygen species generated within mitochondria with insulin resistance in adipose tissue and skeletal muscle, two major insulin sensitive tissues. We outline the relevant mitochondria-derived reactive species, how the mitochondrial redox state is regulated, and methodologies available to measure mitochondrial reactive oxygen species. Importantly, we highlight key experimental issues to be considered when studying the role of mitochondrial reactive oxygen species in insulin resistance. Evaluating the available literature on both mitochondrial reactive oxygen species/redox state and insulin resistance in a variety of biological systems, we conclude that the weight of evidence suggests a likely role for mitochondrial reactive oxygen species in the etiology of insulin resistance in adipose tissue and skeletal muscle. However, major limitations in the methods used to study reactive oxygen species in insulin resistance as well as the lack of data linking mitochondrial reactive oxygen species and cytosolic insulin signaling pathways are significant obstacles in proving the mechanistic link between these two processes. We provide a framework to guide future studies to provide stronger mechanistic information on the link between mitochondrial reactive oxygen species and insulin resistance as understanding the source, localization, nature, and quantity of mitochondrial reactive oxygen species, their targets and downstream signaling pathways may pave the way for important new therapeutic strategies.
Collapse
Affiliation(s)
- Anita Ayer
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Daniel J Fazakerley
- Metabolic Research Laboratory, Wellcome-Medical Research Council Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - David E James
- Charles Perkins Centre, Sydney Medical School, The University of Sydney, Sydney, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia
| | - Roland Stocker
- Heart Research Institute, The University of Sydney, Sydney, New South Wales, Australia; School of Life and Environmental Sciences, The University of Sydney, Sydney, Australia.
| |
Collapse
|
43
|
Frye KA, Sendra KM, Waldron KJ, Kehl-Fie TE. Old dogs, new tricks: New insights into the iron/manganese superoxide dismutase family. J Inorg Biochem 2022; 230:111748. [PMID: 35151099 PMCID: PMC9112591 DOI: 10.1016/j.jinorgbio.2022.111748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 01/14/2022] [Accepted: 01/30/2022] [Indexed: 12/21/2022]
Abstract
Superoxide dismutases (SODs) are ancient enzymes of widespread importance present in all domains of life. Many insights have been gained into these important enzymes over the 50 years since their initial description, but recent studies in the context of microbial pathogenesis have resulted in findings that challenge long established dogmas. The repertoire of SODs that bacterial pathogens encode is diverse both in number and in metal dependencies, including copper, copper and zinc, manganese, iron, and cambialistic enzymes. Other bacteria also possess nickel dependent SODs. Compartmentalization of SODs only partially explains their diversity. The need for pathogens to maintain SOD activity across distinct hostile environments encountered during infection, including those limited for essential metals, is also a driver of repertoire diversity. SOD research using pathogenic microbes has also revealed the apparent biochemical ease with which metal specificity can change within the most common family of SODs. Collectively, these studies are revealing the dynamic nature of SOD evolution, both that of individual SOD enzymes that can change their metal specificity to adapt to fluctuating cellular metal availability, and of a cell's repertoire of SOD isozymes that can be differentially expressed to adapt to fluctuating environmental metal availability in a niche.
Collapse
|
44
|
Antioxidant Role and Cardiolipin Remodeling by Redox-Activated Mitochondrial Ca 2+-Independent Phospholipase A 2γ in the Brain. Antioxidants (Basel) 2022; 11:antiox11020198. [PMID: 35204081 PMCID: PMC8868467 DOI: 10.3390/antiox11020198] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondrial Ca2+-independent phospholipase A2γ (iPLA2γ/PNPLA8) was previously shown to be directly activated by H2O2 and release free fatty acids (FAs) for FA-dependent H+ transport mediated by the adenine nucleotide translocase (ANT) or uncoupling protein 2 (UCP2). The resulting mild mitochondrial uncoupling and consequent partial attenuation of mitochondrial superoxide production lead to an antioxidant effect. However, the antioxidant role of iPLA2γ in the brain is not completely understood. Here, using wild-type and iPLA2γ-KO mice, we demonstrate the ability of tert-butylhydroperoxide (TBHP) to activate iPLA2γ in isolated brain mitochondria, with consequent liberation of FAs and lysophospholipids. The liberated FA caused an increase in respiratory rate, which was fully inhibited by carboxyatractyloside (CATR), a specific inhibitor of ANT. Employing detailed lipidomic analysis, we also demonstrate a typical cleavage pattern for TBHP-activated iPLA2γ, reflecting cleavage of glycerophospholipids from both sn-1 and sn-2 positions releasing saturated FAs, monoenoic FAs, and predominant polyunsaturated FAs. The acute antioxidant role of iPLA2γ-released FAs is supported by monitoring both intramitochondrial superoxide and extramitochondrial H2O2 release. We also show that iPLA2γ-KO mice were more sensitive to stimulation by pro-inflammatory lipopolysaccharide, as reflected by the concomitant increase in protein carbonyls in the brain and pro-inflammatory IL-6 release in the serum. These data support the antioxidant and anti-inflammatory role of iPLA2γ in vivo. Our data also reveal a substantial decrease of several high molecular weight cardiolipin (CL) species and accumulation of low molecular weight CL species in brain mitochondria of iPLA2γ-KO mice. Collectively, our results support a key role of iPLA2γ in the remodeling of lower molecular weight immature cardiolipins with predominantly saturated acyl chains to high molecular weight mature cardiolipins with highly unsaturated PUFA acyl chains, typical for the brain.
Collapse
|
45
|
Radovanovic J, Banjac K, Obradovic M, Isenovic ER. Antioxidant enzymes and vascular diseases. EXPLORATION OF MEDICINE 2021. [DOI: 10.37349/emed.2021.00070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) play a fundamental role in regulating endothelial function and vascular tone in the physiological conditions of a vascular system. However, oxidative stress has detrimental effects on human health, and numerous studies confirmed that high ROS/RNS production contributes to the initiation and progression of cardiovascular diseases. The antioxidant defense has an essential role in the homeostatic functioning of the vascular endothelial system. Endogenous antioxidative defense includes various molecules and enzymes such as superoxide dismutase, catalase, glutathione reductase, and glutathione peroxidase. Together all these antioxidative enzymes are essential for defense against harmful ROS features. ROS are mainly generated from redox-active compounds involved in the mitochondrial respiratory chain. Thus, targeting antioxidative enzymes and mitochondria oxidative balance may be a promising approach for vascular diseases occurrence and treatment. This review summarized the most recent research on the regulation of antioxidative enzymes in vascular diseases.
Collapse
Affiliation(s)
- Jelena Radovanovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| | - Katarina Banjac
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| | - Milan Obradovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| | - Esma R. Isenovic
- Department of Radiobiology and Molecular Genetics, “VINČA” Institute of Nuclear Sciences-National Institute of the Republic of Serbia, University of Belgrade, 522 Belgrade, Serbia
| |
Collapse
|
46
|
Hansen JM, Lucas SM, Ramos CD, Green EJ, Nuttall DJ, Clark DS, Marchant ED, Hancock CR, Piorczynski TB. Valproic acid promotes SOD2 acetylation: A potential mechanism of valproic acid-induced oxidative stress in developing systems. Free Radic Res 2021; 55:1130-1144. [PMID: 34895005 DOI: 10.1080/10715762.2021.2017913] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Valproic acid (VPA) is an antiepileptic, bipolar and migraine medication, which is associated with embryonic dysmorphology, more specifically neural tube defects (NTDs), if taken while pregnant. One mechanism by which VPA may cause NTDs is through oxidative stress that cause disruption of cell signaling. However, mechanisms of VPA-induced oxidative stress are not fully understood. Since VPA is a deacetylase inhibitor, we propose that VPA promotes mitochondrial superoxide dismutase-2 (SOD2) acetylation, decreasing SOD2 activity and increasing oxidant levels. Using the pluripotent embryonal carcinoma cell line, P19, VPA effects were evaluated in undifferentiated and neurodifferentiated cells. VPA treatments increased oxidant levels, oxidized the glutathione (GSH)/glutathione disulfide (GSSG) redox couple, and decreased total SOD and SOD2 activity in undifferentiated P19 cells but not in differentiated P19 cells. VPA caused a specific increase in mitochondrial oxidants in undifferentiated P19 cells, VPA did not alter respirometry measurements. Immunoblot analyses demonstrated that VPA increased acetylation of SOD2 at lysine68 (AcK68 SOD2) in undifferentiated P19 cells but not in differentiated P19 cells. Pretreatments with the Nrf2 inducer, dithiol-3-thione (D3T), in undifferentiated P19 cells prevented increased oxidant levels, GSH/GSSG redox oxidation and restored total SOD and SOD2 activity, correlating with a decrease in AcK68 SOD2 levels. In embryos, VPA decreased total SOD and SOD2 activity and increased levels of AcK68 SOD2, and D3T pretreatments prevented VPA effects, increasing total SOD and SOD2 activity and lowering levels of AcK68 SOD2. These data demonstrate a potential, contributing oxidizing mechanism by which VPA incites teratogenesis in developing systems. Moreover, these data also suggest that Nrf2 interventions may serve as a means to protect developmental signaling and inhibit VPA-induced malformations.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Erik D Marchant
- Department of Nutrition, Dietetics and Food Science, College of Life Sciences, Brigham Young University, Provo, Utah, USA
| | - Chad R Hancock
- Department of Nutrition, Dietetics and Food Science, College of Life Sciences, Brigham Young University, Provo, Utah, USA
| | | |
Collapse
|
47
|
The potential roles of excitatory-inhibitory imbalances and the repressor element-1 silencing transcription factor in aging and aging-associated diseases. Mol Cell Neurosci 2021; 117:103683. [PMID: 34775008 DOI: 10.1016/j.mcn.2021.103683] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/02/2021] [Accepted: 11/06/2021] [Indexed: 12/28/2022] Open
Abstract
Disruptions to the central excitatory-inhibitory (E/I) balance are thought to be related to aging and underlie a host of neural pathologies, including Alzheimer's disease. Aging may induce an increase in excitatory signaling, causing an E/I imbalance, which has been linked to shorter lifespans in mice, flies, and worms. In humans, extended longevity correlates to greater repression of genes involved in excitatory neurotransmission. The repressor element-1 silencing transcription factor (REST) is a master regulator in neural cells and is believed to be upregulated with senescent stimuli, whereupon it counters hyperexcitability, insulin/insulin-like signaling pathway activity, oxidative stress, and neurodegeneration. This review examines the putative mechanisms that distort the E/I balance with aging and neurodegeneration, and the putative roles of REST in maintaining neuronal homeostasis.
Collapse
|
48
|
Tracy EP, Hughes W, Beare JE, Rowe G, Beyer A, LeBlanc AJ. Aging-Induced Impairment of Vascular Function: Mitochondrial Redox Contributions and Physiological/Clinical Implications. Antioxid Redox Signal 2021; 35:974-1015. [PMID: 34314229 PMCID: PMC8905248 DOI: 10.1089/ars.2021.0031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: The vasculature responds to the respiratory needs of tissue by modulating luminal diameter through smooth muscle constriction or relaxation. Coronary perfusion, diastolic function, and coronary flow reserve are drastically reduced with aging. This loss of blood flow contributes to and exacerbates pathological processes such as angina pectoris, atherosclerosis, and coronary artery and microvascular disease. Recent Advances: Increased attention has recently been given to defining mechanisms behind aging-mediated loss of vascular function and development of therapeutic strategies to restore youthful vascular responsiveness. The ultimate goal aims at providing new avenues for symptom management, reversal of tissue damage, and preventing or delaying of aging-induced vascular damage and dysfunction in the first place. Critical Issues: Our major objective is to describe how aging-associated mitochondrial dysfunction contributes to endothelial and smooth muscle dysfunction via dysregulated reactive oxygen species production, the clinical impact of this phenomenon, and to discuss emerging therapeutic strategies. Pathological changes in regulation of mitochondrial oxidative and nitrosative balance (Section 1) and mitochondrial dynamics of fission/fusion (Section 2) have widespread effects on the mechanisms underlying the ability of the vasculature to relax, leading to hyperconstriction with aging. We will focus on flow-mediated dilation, endothelial hyperpolarizing factors (Sections 3 and 4), and adrenergic receptors (Section 5), as outlined in Figure 1. The clinical implications of these changes on major adverse cardiac events and mortality are described (Section 6). Future Directions: We discuss antioxidative therapeutic strategies currently in development to restore mitochondrial redox homeostasis and subsequently vascular function and evaluate their potential clinical impact (Section 7). Antioxid. Redox Signal. 35, 974-1015.
Collapse
Affiliation(s)
- Evan Paul Tracy
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - William Hughes
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Jason E Beare
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA.,Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, Kentucky, USA
| | - Gabrielle Rowe
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA
| | - Andreas Beyer
- Department of Medicine and Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Amanda Jo LeBlanc
- Department of Physiology, University of Louisville, Louisville, Kentucky, USA.,Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| |
Collapse
|
49
|
Fernando LM, Adeel S, Basar MA, Allen AK, Duttaroy A. In-gel SOD assay reveals SOD-2 is the single active, water-soluble SOD enzyme in C. elegans. Free Radic Res 2021; 55:619-624. [PMID: 34514925 DOI: 10.1080/10715762.2021.1979228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The nematode C. elegans has a contingent of five sod genes, one of the largest among aerobic organisms. Earlier studies revealed each of the five sod genes is capable of making perfectly active SOD proteins in heterologous expression systems therefore none appears to be a pseudogene. Yet deletion of the entire contingent of sod genes fails to impose any effect on the survival of C. elegans except these animals appear more sensitive to extraneously applied oxidative stress conditions. We asked how many of the five sod genes are actually making active SOD enzymes in C. elegans through the usage of in-gel SOD activity analysis and by using KCN as a selective inhibitor against Cu-ZnSOD enzyme(s). Here we provide evidence that out of the five SOD proteins only the mitochondrial SOD is active in the water-soluble fraction of C. elegans extracts albeit at an apparently much lower activity than the multiple active SODs in D. melanogaster and E. coli. We had no opportunity to test the activity of Sod-4a isoform which is possibly a membrane-bound form of SOD. The mutant analysis further confirmed that among the two mitochondrial SOD proteins, SOD-2 is the only naturally active SOD in C. elegans.
Collapse
Affiliation(s)
- Lourds M Fernando
- Department of Biology, Howard University, Washington, DC, USA.,St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Silwat Adeel
- Department of Biology, Howard University, Washington, DC, USA
| | - Mohammed Abul Basar
- Department of Biology, Howard University, Washington, DC, USA.,Stem Cell Biochemistry Unit, National Institute of Dental and Craniofacial Research National Institutes of Health, Bethesda, MD, USA
| | - Anna K Allen
- Department of Biology, Howard University, Washington, DC, USA
| | - Atanu Duttaroy
- Department of Biology, Howard University, Washington, DC, USA
| |
Collapse
|
50
|
Mapuskar KA, Steinbach EJ, Zaher A, Riley DP, Beardsley RA, Keene JL, Holmlund JT, Anderson CM, Zepeda-Orozco D, Buatti JM, Spitz DR, Allen BG. Mitochondrial Superoxide Dismutase in Cisplatin-Induced Kidney Injury. Antioxidants (Basel) 2021; 10:antiox10091329. [PMID: 34572961 PMCID: PMC8469643 DOI: 10.3390/antiox10091329] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 02/06/2023] Open
Abstract
Cisplatin is a chemotherapy agent commonly used to treat a wide variety of cancers. Despite the potential for both severe acute and chronic side effects, it remains a preferred therapeutic option for many malignancies due to its potent anti-tumor activity. Common cisplatin-associated side-effects include acute kidney injury (AKI) and chronic kidney disease (CKD). These renal injuries may cause delays and potentially cessation of cisplatin therapy and have long-term effects on renal function reserve. Thus, developing mechanism-based interventional strategies that minimize cisplatin-associated kidney injury without reducing efficacy would be of great benefit. In addition to its action of cross-linking DNA, cisplatin has been shown to affect mitochondrial metabolism, resulting in mitochondrially derived reactive oxygen species (ROS). Increased ROS formation in renal proximal convoluted tubule cells is associated with cisplatin-induced AKI and CKD. We review the mechanisms by which cisplatin may induce AKI and CKD and discuss the potential of mitochondrial superoxide dismutase mimetics to prevent platinum-associated nephrotoxicity.
Collapse
Affiliation(s)
- Kranti A. Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Emily J. Steinbach
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Amira Zaher
- Biomedical Science Program, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA;
| | - Dennis P. Riley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Robert A. Beardsley
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jeffery L. Keene
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Jon T. Holmlund
- Galera Therapeutics, Inc., Malvern, PA 19355, USA; (D.P.R.); (R.A.B.); (J.L.K.); (J.T.H.)
| | - Carryn M. Anderson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Diana Zepeda-Orozco
- Center for Clinical and Translational Research, The Abigail Wexner Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA;
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Nephrology, Department of Pediatrics, Nationwide Children’s Hospital, Columbus, OH 43205, USA
| | - John M. Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa, Iowa City, IA 52242, USA; (K.A.M.); (E.J.S.); (C.M.A.); (J.M.B.); (D.R.S.)
- Correspondence: ; Tel.: +1-319-335-8019; Fax: +1-319-335-8039
| |
Collapse
|