1
|
Zheng J, Manabe Y, Sugawara T. Preventive effect of siphonaxanthin, a carotenoid from green algae, against diabetic nephropathy and lipid metabolism insufficiency in skeletal muscle. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159604. [PMID: 39986648 DOI: 10.1016/j.bbalip.2025.159604] [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: 10/29/2024] [Revised: 02/07/2025] [Accepted: 02/17/2025] [Indexed: 02/24/2025]
Abstract
Diabetic nephropathy is a complication of diabetes mellitus characterized by the gradual progression of renal insufficiency, resulting in renal failure. Approximately 15 % or more of patients with type 2 diabetes mellitus have diabetic nephropathy. Siphonaxanthin is a green algal carotenoid noted for its strong biological activities, including anti-obesity effects. In this study, we aimed to evaluate the preventive effects of siphonaxanthin on diabetic nephropathy using db/db mice as a type 2 diabetes mellitus and diabetic nephropathy model. Ingestion of AIN-93G containing 0.004 % w/w siphonaxanthin did not improve plasma creatinine and urine albumin levels but significantly mitigated renal morphological changes in diabetic mice. Moreover, siphonaxanthin restored the decreased mRNA expression of fatty acid β-oxidation-related proteins in the skeletal muscle. These results indicate that siphonaxanthin can potentially ameliorate type 2 diabetes mellitus-induced kidney damage and lipid metabolism insufficiency in skeletal muscle. This study provides a possible daily nutraceutical solution for treating diabetic nephropathy and lipid metabolic abnormalities.
Collapse
Affiliation(s)
- Jiawen Zheng
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University. Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan
| | - Yuki Manabe
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University. Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan.
| | - Tatsuya Sugawara
- Division of Applied Biosciences, Graduate School of Agriculture, Kyoto University. Kitashirakawa Oiwake-cho, Sakyo-ku, Kyoto 606-8502, Japan.
| |
Collapse
|
2
|
Kyriakopoulou E, van Kampen SJ, Wehrens M, Han SJ, de Ruiter H, Monshouwer-Kloots J, Marshall E, Brodehl A, van der Kraak P, te Riele AS, van Aarnhem EE, van Laake LW, Tsui H, Boogerd CJ, van Rooij E. EPAS1 induction drives myocardial degeneration in desmoplakin-cardiomyopathy. iScience 2025; 28:111895. [PMID: 40034852 PMCID: PMC11872638 DOI: 10.1016/j.isci.2025.111895] [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: 08/22/2024] [Revised: 11/24/2024] [Accepted: 01/22/2025] [Indexed: 03/05/2025] Open
Abstract
Arrhythmogenic cardiomyopathy (ACM) is frequently attributed to desmosomal mutations, such as those in the desmoplakin (DSP) gene. Patients with DSP-cardiomyopathy are predisposed to myocardial degeneration and arrhythmias. Despite advancements, the underlying molecular mechanisms remain incompletely understood, thus limiting therapeutic options. Here, we employed spatial transcriptomics on an explanted heart from a patient with a pathogenic DSP variant. Our transcriptional analysis revealed endothelial PAS domain-containing protein 1 (EPAS1) as a potential regulator of mitochondrial homeostasis in stressed cardiomyocytes. Elevated EPAS1 levels were associated with mitochondrial dysfunction and hypoxic stress in both human-relevant in vitro ACM models and additional explanted hearts with genetic cardiomyopathy. Collectively, cardiomyocytes bearing pathogenic DSP variants exhibit mitochondrial dysfunction, increased apoptosis, and impaired contractility, which are linked to the increased EPAS1 levels. These findings implicate EPAS1 as a key regulator of myocardial degeneration in DSP-cardiomyopathy, which expand to other forms of ACM.
Collapse
Affiliation(s)
- Eirini Kyriakopoulou
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Sebastiaan J. van Kampen
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Martijn Wehrens
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Su Ji Han
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hesther de Ruiter
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jantine Monshouwer-Kloots
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Emma Marshall
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Andreas Brodehl
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Bad Oeynhausen, Germany
| | - Petra van der Kraak
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Anneline S.J.M. te Riele
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
- Netherlands Heart Institute, Utrecht, the Netherlands
| | - Egidius E.H.L. van Aarnhem
- Division of Heart and Lungs, Department of Cardiothoracic Surgery, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Linda W. van Laake
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Hoyee Tsui
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Cornelis J. Boogerd
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
| | - Eva van Rooij
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences (KNAW) and University Medical Center Utrecht, Utrecht, the Netherlands
- Division Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
3
|
Trambas IA, Bowen L, Thallas-Bonke V, Snelson M, Sourris KC, Laskowski A, Tauc M, Rubera I, Zheng G, Harris DCH, Kantharidis P, Shimizu T, Cooper ME, Tan SM, Coughlan MT. Proximal tubular deletion of superoxide dismutase-2 reveals disparate effects on kidney function in diabetes. Redox Biol 2025; 82:103601. [PMID: 40127616 DOI: 10.1016/j.redox.2025.103601] [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: 12/18/2024] [Revised: 03/09/2025] [Accepted: 03/17/2025] [Indexed: 03/26/2025] Open
Abstract
There is a large body of evidence implicating mitochondrial reactive oxygen species (ROS) overproduction and oxidative stress in the development of diabetic kidney disease and the deficiency of mitochondrial antioxidant systems in the kidney, such as manganese superoxide dismutase (MnSOD/SOD2) have been identified. The proximal tubules of the kidney are densely packed with mitochondria thereby providing energy via oxidative phosphorylation in order to drive active transport for proximal tubular reabsorption of solutes from the glomerular filtrate. We hypothesized that maintenance of MnSOD function in the proximal tubules would be critical to maintain kidney health in diabetes. Here, we induced targeted deletion of SOD2 in the proximal tubules of the kidney in Ins2Akita diabetic mice (SODptKO mice) and show that 20 weeks of SOD2 deletion leads to no major impairment of kidney function and structure, despite these mice displaying enhanced albuminuria and kidney lipid peroxidation (8-isoprostanes). Plasma cystatin C, which is a surrogate marker of glomerular filtration was not altered in SODptKO diabetic mice and histological assessment of the kidney cortex revealed no change in kidney fibrosis. Thus, our findings suggest that deletion of SOD2 in the proximal tubular compartment of the kidney induces a more subtle phenotype than expected, shedding light on the involvement of SOD2 and the proximal tubular compartment in the pathogenesis of diabetic kidney disease.
Collapse
Affiliation(s)
- Inez A Trambas
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Lilliana Bowen
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Vicki Thallas-Bonke
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Matthew Snelson
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Karly C Sourris
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Adrienne Laskowski
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Michel Tauc
- Laboratoire de Physiomédecine Moléculaire, Université Côte D'Azur, CNRS, LP2M, 7370, Nice Cedex 2, France
| | - Isabelle Rubera
- Laboratoire de Physiomédecine Moléculaire, Université Côte D'Azur, CNRS, LP2M, 7370, Nice Cedex 2, France
| | - Guoping Zheng
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, 2145, Australia
| | - David C H Harris
- Centre for Transplantation and Renal Research, Westmead Institute for Medical Research, University of Sydney, Sydney, NSW, 2145, Australia
| | - Phillip Kantharidis
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Takahiko Shimizu
- Department of Food and Reproductive Function Advanced Research, Juntendo University Graduate School of Medicine, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Mark E Cooper
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Sih Min Tan
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia
| | - Melinda T Coughlan
- Department of Diabetes, School of Translational Medicine, Monash University, Melbourne, 3004, Victoria, Australia; Baker Heart and Diabetes Institute, Melbourne, 3004, Victoria, Australia; Drug Discovery Biology, Monash Institute of Pharmaceutical Science, Monash University Parkville Campus, 381 Royal Parade, Parkville, 3052, Victoria, Australia.
| |
Collapse
|
4
|
Schell JR, Wei SJ, Zhang J, Trevino R, Chen WH, Aguilar L, Qian W, Corbett CW, Jiang H, Dong FF, Chocron ES, Nazarullah A, Chang J, Flanagan ME, Glatt V, Dikalov S, Munkácsy E, Horikoshi N, Gius D. MnSOD non-acetylation mimic knock-in mice exhibit dilated cardiomyopathy. Free Radic Biol Med 2025; 229:58-67. [PMID: 39824446 DOI: 10.1016/j.freeradbiomed.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/30/2024] [Accepted: 01/13/2025] [Indexed: 01/20/2025]
Abstract
Manganese superoxide dismutase (MnSOD/SOD2) is an essential mitochondrial enzyme that detoxifies superoxide radicals generated during oxidative respiration. MnSOD/SOD2 lysine 68 acetylation (K68-Ac) is an important post-translational modification (PTM) that regulates enzymatic activity, responding to nutrient status or oxidative stress, and elevated levels have been associated with human illness. To determine the in vivo role of MnSOD-K68 in the heart, we used a whole-body non-acetylation mimic mutant (MnSODK68R) knock-in mouse. These mice exhibited several cardiovascular phenotypes, including lower blood pressure, decreased ejection fraction, and importantly, dilated cardiomyopathy, as evidenced by echocardiography at four months of age. In addition, both mouse embryo fibroblasts (MEFs) and cardiovascular tissue from MnSODK68R/K68R mice exhibited an increase in cellular senescence. Finally, MnSODK68R/K68R mouse hearts also showed an increase in lipid peroxidation. We conclude that constitutively active MnSOD detoxification activity, lacking the normal switch between non-acetylated and acetylated forms, dysregulates mitochondrial physiology during development, leading to dilated cardiomyopathy.
Collapse
Affiliation(s)
- Joseph R Schell
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Sung-Jen Wei
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Jun Zhang
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Rolando Trevino
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Wan Hsi Chen
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Leonardo Aguilar
- Department of Orthopedic Surgery, UT Health San Antonio, TX, USA
| | - Wei Qian
- Houston Methodist Cancer Center, Houston, TX, USA; Houston Methodist Research Institute, Houston, TX, USA
| | - Cole W Corbett
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, TX, USA
| | - Haiyan Jiang
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Felix F Dong
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - E Sandra Chocron
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Alia Nazarullah
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Department of Pathology, UT Health San Antonio, TX, USA
| | - Jenny Chang
- Houston Methodist Cancer Center, Houston, TX, USA; Houston Methodist Research Institute, Houston, TX, USA
| | - Margaret E Flanagan
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UT Health San Antonio, TX, USA; Department of Pathology, UT Health San Antonio, TX, USA
| | - Vaida Glatt
- Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA; Department of Orthopedic Surgery, UT Health San Antonio, TX, USA
| | - Sergey Dikalov
- Vanderbilt University Medical Center, Vanderbilt University, Nashville, TN, USA
| | - Erin Munkácsy
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - Nobuo Horikoshi
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA; Barshop Institute for Longevity and Aging Studies, UT Health San Antonio, TX, USA
| | - David Gius
- Department of Radiation Oncology, Mays Cancer Center at UT Health San Antonio MD Anderson, Joe R. and Teresa Lozano Long School of Medicine, TX, USA.
| |
Collapse
|
5
|
Fu R, Guo J, Jin H, Li Z, Xia J, Tang Y, Ding Y, Zhao H, Ye D, Xu J. Boosting Enzyme-Like Activity Through Controlled Coordination of Metal Single-Atoms in Rhombic Cavity of Graphyne. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409113. [PMID: 39718224 DOI: 10.1002/smll.202409113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/22/2024] [Indexed: 12/25/2024]
Abstract
The coordination number of metal single-atoms, being an important factor, should be precisely controlled as it affects both activity and specificity. Herein, the introduction of nitrogen into the para position of benzene in graphyne (2N-GY) is proposed as a carrier for anchoring metal single-atoms onto pyrazine nitrogen. These atoms are then coordinated to the center of the rhombic cavity, ensuring precisely controlled coordination numbers. Inspired by natural enzymes, a series of transition metal single-atom nanozymes (SANs) are constructed utilizing 2N-GY. Both theory calculations and experiments demonstrated that the incorporation of metal single-atoms of Fe, Mn, and Mo effectively augmented the enzyme-like activity, with Mo/GY exhibiting the highest peroxidase -like activity. Based on the differing activities of M/GY (M = Fe, Mn, Mo) and the varying abilities of sulfhydryl groups in biothiols to occupy metal active sites, sensing arrays for the high-throughput identification of six biothiols are constructed. After combining the fingerprint feature responses of biothiols and SANs with machine learning algorithms, the efficient distinction of the six biothiols is achieved. This study introduces a novel pathway for designing and synthesizing SANs that mimic natural enzymes.
Collapse
Affiliation(s)
- Ruixue Fu
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Guo
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Huan Jin
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
- Shanxi Inspection and Testing Center, Taiyuan, Shanxi, 030006, P. R. China
| | - Zhen Li
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Jianing Xia
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Ya Tang
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Yaping Ding
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Hongbin Zhao
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Daixin Ye
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| | - Jiaqiang Xu
- Department of Chemistry & Institute for Sustainable Energy, College of Sciences, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
6
|
Abe H, Indo HP, Ito H, Majima HJ, Tanaka T. Synephrine Inhibits Oxidative Stress and H 2O 2-Induced Premature Senescence. Cell Biochem Biophys 2025:10.1007/s12013-025-01669-7. [PMID: 39832117 DOI: 10.1007/s12013-025-01669-7] [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] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Synephrine, a protoalkaloid found in Citrus aurantium (CA) peels, exerts lipolytic, anti-inflammatory, and vasoconstrictive effects; however, its antioxidant activity remains unclear. In this study, electron spin resonance spectroscopy revealed that synephrine scavenged both hydroxyl and superoxide anion radicals. Several external stimuli, such as H2O2, X-rays, and ultraviolet (UV) radiation, cause stress-induced premature senescence (SIPS). As oxidative stress induces SIPS, we hypothesized that synephrine, an antioxidant, would suppress H2O2-induced premature senescence in WI-38 cells. Synephrine significantly decreased the reactive oxygen species levels induced by H2O2, thereby reducing lipid peroxidation, and oxidative DNA damage and preventing SIPS. Additionally, synephrine inhibited mitochondrial dysfunction in H2O2-treated WI-38 cells. The expression levels of p53, p21, and p16-INK4A, which are involved in the induction of cell cycle arrest in SIPS, were significantly lower in synephrine-treated cells than in untreated cells. Our results indicate that synephrine inhibits H2O2-induced oxidative stress and mitochondrial dysfunction, suppressing premature senescence by inhibiting activation of the p53-p21 and p16-INK4A-pRB pathways.
Collapse
Affiliation(s)
- Hiroshi Abe
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
| | - Hiroko P Indo
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan.
| | - Hiromu Ito
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
- Quantum RedOx Chemistry Team, Quantum Life Spin Group, Institute for Quantum Life Science (iQLS), National Institutes for Quantum Science and Technology (QST), Chiba, 263-8555, Japan
| | - Hideyuki J Majima
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Tatsuro Tanaka
- Department of Maxillofacial Radiology, Field of Oncology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, 890-8544, Japan
| |
Collapse
|
7
|
Lutsenko S, Roy S, Tsvetkov P. Mammalian copper homeostasis: physiological roles and molecular mechanisms. Physiol Rev 2025; 105:441-491. [PMID: 39172219 PMCID: PMC11918410 DOI: 10.1152/physrev.00011.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/15/2024] [Accepted: 08/18/2024] [Indexed: 08/23/2024] Open
Abstract
In the past decade, evidence for the numerous roles of copper (Cu) in mammalian physiology has grown exponentially. The discoveries of Cu involvement in cell signaling, autophagy, cell motility, differentiation, and regulated cell death (cuproptosis) have markedly extended the list of already known functions of Cu, such as a cofactor of essential metabolic enzymes, a protein structural component, and a regulator of protein trafficking. Novel and unexpected functions of Cu transporting proteins and enzymes have been identified, and new disorders of Cu homeostasis have been described. Significant progress has been made in the mechanistic studies of two classic disorders of Cu metabolism, Menkes disease and Wilson's disease, which paved the way for novel approaches to their treatment. The discovery of cuproptosis and the role of Cu in cell metastatic growth have markedly increased interest in targeting Cu homeostatic pathways to treat cancer. In this review, we summarize the established concepts in the field of mammalian Cu physiology and discuss how new discoveries of the past decade expand and modify these concepts. The roles of Cu in brain metabolism and in cell functional speciation and a recently discovered regulated cell death have attracted significant attention and are highlighted in this review.
Collapse
Affiliation(s)
- Svetlana Lutsenko
- Department of Physiology, Johns Hopkins Medical Institutes, Baltimore, Maryland, United States
| | - Shubhrajit Roy
- Department of Physiology, Johns Hopkins Medical Institutes, Baltimore, Maryland, United States
| | - Peter Tsvetkov
- Department of Pathology, Cancer Center, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States
| |
Collapse
|
8
|
Nguyen HVM, Ran Q, Salmon AB, Bumsoo A, Chiao YA, Bhaskaran S, Richardson A. Mouse models used to test the role of reactive oxygen species in aging and age-related chronic diseases. Free Radic Biol Med 2024; 225:617-629. [PMID: 39419456 PMCID: PMC11624111 DOI: 10.1016/j.freeradbiomed.2024.10.269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/13/2024] [Accepted: 10/02/2024] [Indexed: 10/19/2024]
Abstract
With the development of the technology to generate transgenic and knockout mice in the 1990s, investigators had a powerful tool to directly test the impact of altering a specific gene on a biological process or disease. Over the past three decades, investigators have used transgenic and knockout mouse models, which have altered expression of antioxidant genes, to test the role of oxidative stress/damage in aging and age-related diseases. In this comprehensive review, we describe the studies using transgenic and knockout mouse models to test the role of oxidative stress/damage in aging (longevity) and three age-related diseases, e.g., sarcopenia, cardiac aging, and Alzheimer's Disease. While longevity was consistently altered only by one transgenic and one knockout mouse model as predicted by the Oxidative Stress Theory of Aging, the incidence/progression of the three age-related diseases (especially Alzheimer's disease) were robustly impacted when the expression of various antioxidant genes was altered using transgenic and knockout mouse models.
Collapse
Affiliation(s)
- Hoang Van M Nguyen
- Department of Nutritional Sciences, University of Oklahoma Health Sciences, Oklahoma City, OK, USA
| | - Qitao Ran
- Department of Cell Systems & Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; VA South Texas Health Care System, San Antonio, TX, USA
| | - Adam B Salmon
- Department of Molecular Medicine, Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; VA South Texas Health Care System, San Antonio, TX, USA
| | - Ahn Bumsoo
- Gerontology and Geriatric Medicine, Wake Forest University School of Medicine, Winston-Salem, North Carolina, USA
| | - Ying Ann Chiao
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Shylesh Bhaskaran
- Aging & Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Arlan Richardson
- Department of Biochemistry & Molecular Biology, University of Oklahoma Health Sciences, Oklahoma City, OK, USA; VA Oklahoma Health Care System, Oklahoma City, OK, USA.
| |
Collapse
|
9
|
Monteiro KKAC, Damous LL, Shiroma ME, Termini L, Cipolla-Neto J, Simões RDS, da Silva RF, Turri JA, Baracat EC, Soares-Junior JM. Melatonin increases superoxide dismutase 2 (SOD2) levels and improves rat ovarian graft function after transplantation. J Ovarian Res 2024; 17:204. [PMID: 39415169 PMCID: PMC11481372 DOI: 10.1186/s13048-024-01512-2] [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: 07/03/2024] [Accepted: 09/07/2024] [Indexed: 10/18/2024] Open
Abstract
BACKGROUND Ovarian cryopreservation is a promising technique despite being hindered by damage from freezing and thawing. Melatonin can mitigate this outcome. OBJECTIVE This study aimed to evaluate the effect of melatonin on the follicular dynamics of ovarian tissue in a cryopreserved cell culture. METHODS Three-month-old adult female Wistar rats (n = 24) weighing approximately 250 g were oophorectomized and divided into two groups (n = 12): the control group (CG) and the melatonin group (MG). In the CG, slow cryopreservation was performed according to the standard protocol with Medium M2 and dimethyl sulfoxide (DMSO). In MG, melatonin diluted in ethyl alcohol vehicle at a concentration of 0.1 μm was added to the culture medium. In both groups, the ovaries were cryopreserved by slow freezing and kept in liquid nitrogen for 24 h. Subsequently, after thawing, the ovaries were reimplanted in the retroperitoneum, one on each side of the great vessels (inferior vena cava and aorta). After 30 days, the animals were euthanized during the diestrus phase; then, the grafts were removed and processed for histomorphometric and immunohistochemical analyses, whereas the blood was subjected to biochemical analysis. Student's t test was used to assess the difference between the groups. RESULTS The FSH levels in MG (83.79 ± 32.37) were lower than those in CG (120.52 ± 36.59), p = 0.03. The FSH/AMH ratios were also lower in MG (3.53 ± 1.13) than in CG (6.52 ± 2.85), p = 0.001. The SOD2 immunoexpression was higher in MG than in CG regarding all parameters except for the degenerated follicles (follicular cells and internal thecal cells): CG (16.80 ± 4.80 [13.36-20.24]) and MG (14.91 ± 4.04 [12.01-17.79]) p = 0.351. Statistically, the difference in intact follicles (theca + interstitium) between CG (6.60 ± 2.59 [4.75-8.45]) and MG (9.31 ± 3.09 [7.09-11.51]) was significant (p = 0.049), with a small difference in the expression of regular antral follicles. CONCLUSIONS Melatonin can improve the quality of cryopreserved tissues, as evidenced in this study, and the evaluation of cryopreserved ovarian grafts, as shown in the melatonin group with better hormonal parameters and greater immunohistochemical expression of the SOD2 antioxidant. Thus, damage is reduced during cryopreservation and transplantation is improved.
Collapse
Affiliation(s)
- Karla Krislane Alves Costa Monteiro
- Department of Obstetrics and Gynecology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, 05403-010, Brazil.
| | - Luciana Lamarão Damous
- Department of Obstetrics and Gynecology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, 05403-010, Brazil
| | - Marcos Eiji Shiroma
- Department of Obstetrics and Gynecology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, 05403-010, Brazil
| | - Lara Termini
- Cancer Innovation Laboratory, Center for Translational Research in Oncology (CTO), Cancer Institute of the State of São Paulo (ICESP), Faculdade de Medicina da Universidade de São Paulo, São Paulo, 01246-000, Brazil
| | - José Cipolla-Neto
- Institute of Biomedical Sciences (IBC-I), Neurobiology Laboratory, Physiology Department, Universidade de São Paulo, São Paulo, 05508-000, Brazil
| | - Ricardo Dos Santos Simões
- Department of Obstetrics and Gynecology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, 05403-010, Brazil
| | - Rinaldo Florencio da Silva
- Department of Morphology and Genetics/Discipline of Histology and Structural Biology, Escola Paulista de Medicina - Universidade Federal de São Paulo (EPM- UNIFESP), São Paulo, 04023-062, Brazil
| | - José Antonio Turri
- Department of Obstetrics and Gynecology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, 05403-010, Brazil
| | - Edmund C Baracat
- Department of Obstetrics and Gynecology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, 05403-010, Brazil
| | - Jose Maria Soares-Junior
- Department of Obstetrics and Gynecology, Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo (HCFMUSP), São Paulo, 05403-010, Brazil
| |
Collapse
|
10
|
Anwar I, Wang X, Pratt RE, Dzau VJ, Hodgkinson CP. The impact of aging on cardiac repair and regeneration. J Biol Chem 2024; 300:107682. [PMID: 39159819 PMCID: PMC11414664 DOI: 10.1016/j.jbc.2024.107682] [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: 04/29/2024] [Revised: 07/10/2024] [Accepted: 08/02/2024] [Indexed: 08/21/2024] Open
Abstract
In contrast to neonates and lower organisms, the adult mammalian heart lacks any capacity to regenerate following injury. The vast majority of our understanding of cardiac regeneration is based on research in young animals. Research in aged individuals is rare. This is unfortunate as aging induces many changes in the heart. The first part of this review covers the main technologies being pursued in the cardiac regeneration field and how they are impacted by the aging processes. The second part of the review covers the significant amount of aging-related research that could be used to aid cardiac regeneration. Finally, a perspective is provided to suggest how cardiac regenerative technologies can be improved by addressing aging-related effects.
Collapse
Affiliation(s)
- Iqra Anwar
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Xinghua Wang
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Richard E Pratt
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Victor J Dzau
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA
| | - Conrad P Hodgkinson
- Mandel Center for Heart and Vascular Research, Duke Cardiovascular Research Center, Duke University Medical Center, Durham, North Carolina, USA.
| |
Collapse
|
11
|
Shi C, Wen Z, Yang Y, Shi L, Liu D. NAD+ metabolism and therapeutic strategies in cardiovascular diseases. ATHEROSCLEROSIS PLUS 2024; 57:1-12. [PMID: 38974325 PMCID: PMC11223091 DOI: 10.1016/j.athplu.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/25/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024]
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a central and pleiotropic metabolite involved in cellular energy metabolism, cell signaling, DNA repair, and protein modifications. Cardiovascular diseases (CVDs) are the leading cause of death worldwide. Metabolic stress and aging directly affect the cardiovascular system. Compelling data suggest that NAD + levels decrease with age, obesity, and hypertension, which are all notable risk factors for CVD. In addition, the therapeutic elevation of NAD + levels reduces chronic low-grade inflammation, reactivates autophagy and mitochondrial biogenesis, and enhances oxidative metabolism in vascular cells of humans and rodents with vascular disorders. In preclinical models, NAD + boosting can also expand the health span, prevent metabolic syndrome, and decrease blood pressure. Moreover, NAD + storage by genetic, pharmacological, or natural dietary NAD + -increasing strategies has recently been shown to be effective in improving the pathophysiology of cardiac and vascular health in different animal models, and human health. Here, we review and discuss NAD + -related mechanisms pivotal for vascular health and summarize recent experimental evidence in NAD + research directly related to vascular disease, including atherosclerosis, and coronary artery disease. Finally, we comparatively assess distinct NAD + precursors for their clinical efficacy and the efficiency of NAD + elevation in the treatment of major CVD. These findings may provide ideas for new therapeutic strategies to prevent and treat CVD in the clinic.
Collapse
Affiliation(s)
- Chongxu Shi
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
| | - Zhaozhi Wen
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
| | - Yihang Yang
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
| | - Linsheng Shi
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Dong Liu
- Nantong Laboratory of Development and Diseases, School of Life Science, Nantong University, Nantong, China
- Department of Cardiology, The Second Affiliated Hospital of Nantong University, Nantong, China
- Co-Innovation Center of Neuroregeneration, Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Nantong, China
| |
Collapse
|
12
|
Lin F, Liang X, Meng Y, Zhu Y, Li C, Zhou X, Hu S, Yi N, Lin Q, He S, Sun Y, Sheng J, Fan H, Li L, Peng L. Unmasking Protein Phosphatase 2A Regulatory Subunit B as a Crucial Factor in the Progression of Dilated Cardiomyopathy. Biomedicines 2024; 12:1887. [PMID: 39200351 PMCID: PMC11352103 DOI: 10.3390/biomedicines12081887] [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: 07/23/2024] [Revised: 08/06/2024] [Accepted: 08/15/2024] [Indexed: 09/02/2024] Open
Abstract
Dilated cardiomyopathy (DCM) is one of the major causes of heart failure. Although significant progress has been made in elucidating the underlying mechanisms, further investigation is required for clarifying molecular diagnostic and therapeutic targets. In this study, we found that the mRNA level of protein phosphatase 2 regulatory subunit B' delta (Ppp2r5d) was altered in the peripheral blood plasma of DCM patients. Knockdown of Ppp2r5d in murine cardiomyocytes increased the intracellular levels of reactive oxygen species (ROS) and inhibited adenosine triphosphate (ATP) synthesis. In vivo knockdown of Ppp2r5d in an isoproterenol (ISO)-induced DCM mouse model aggravated the pathogenesis and ultimately led to heart failure. Mechanistically, Ppp2r5d-deficient cardiomyocytes showed an increase in phosphorylation of STAT3 at Y705 and a decrease in phosphorylation of STAT3 at S727. The elevated levels of phosphorylation at Y705 in STAT3 triggered the upregulation of interleukin 6 (IL6) expression. Moreover, the decreased phosphorylation at S727 in STAT3 disrupted mitochondrial electron transport chain function and dysregulated ATP synthesis and ROS levels. These results hereby reveal a novel role for Ppp2r5d in modulating STAT3 pathway in DCM, suggesting it as a potential target for the therapy of the disease.
Collapse
Affiliation(s)
- Fang Lin
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Xiaoting Liang
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University School of Life Sciences and Technology, Tongji University, Shanghai 200120, China
| | - Yilei Meng
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Yuping Zhu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Chenyu Li
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Xiaohui Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Sangyu Hu
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Na Yi
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Qin Lin
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Siyu He
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Yizhuo Sun
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Jie Sheng
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Huimin Fan
- Research Center for Translational Medicine, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Shanghai Heart Failure Research Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Li Li
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| | - Luying Peng
- State Key Laboratory of Cardiology and Medical Innovation Center, Shanghai East Hospital, Tongji University School of Medicine, Tongji University, Shanghai 200120, China; (F.L.)
- Laboratory of Molecular Genetics and Stem Cell Differentiation, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
- Department of Cell and Genetics, Tongji University School of Medicine, Tongji University, Shanghai 200120, China
| |
Collapse
|
13
|
Yan S, Lu J, Chen B, Yuan L, Chen L, Ju L, Cai W, Wu J. The Multifaceted Role of Alpha-Lipoic Acid in Cancer Prevention, Occurrence, and Treatment. Antioxidants (Basel) 2024; 13:897. [PMID: 39199143 PMCID: PMC11351715 DOI: 10.3390/antiox13080897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024] Open
Abstract
Alpha-lipoic acid (ALA) is a naturally occurring compound synthesized by mitochondria and widely distributed in both animal and plant tissues. It primarily influences cellular metabolism and oxidative stress networks through its antioxidant properties and is an important drug for treating metabolic diseases associated with oxidative damage. Nevertheless, research indicates that the mechanism by which ALA affects cancer cells is distinct from that observed in normal cells, exhibiting pro-oxidative properties. Therefore, this review aims to describe the main chemical and biological functions of ALA in the cancer environment, including its mechanisms and effects in tumor prevention and anticancer activity, as well as its role as an adjunctive drug in cancer therapy. We specifically focus on the interactions between ALA and various carcinogenic and anti-carcinogenic pathways and discuss ALA's pro-oxidative capabilities in the unique redox environment of cancer cells. Additionally, we elaborate on ALA's roles in nanomedicine, hypoxia-inducible factors, and cancer stem cell research, proposing hypotheses and potential explanations for currently unresolved issues.
Collapse
Affiliation(s)
- Shuai Yan
- Medical School, Nantong University, Nantong 226300, China; (S.Y.); (J.L.); (B.C.)
| | - Jiajie Lu
- Medical School, Nantong University, Nantong 226300, China; (S.Y.); (J.L.); (B.C.)
| | - Bingqing Chen
- Medical School, Nantong University, Nantong 226300, China; (S.Y.); (J.L.); (B.C.)
| | - Liuxia Yuan
- Institute of Liver Diseases, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226300, China; (L.Y.); (L.C.); (L.J.)
| | - Lin Chen
- Institute of Liver Diseases, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226300, China; (L.Y.); (L.C.); (L.J.)
| | - Linglin Ju
- Institute of Liver Diseases, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226300, China; (L.Y.); (L.C.); (L.J.)
| | - Weihua Cai
- Department of Hepatobiliary Surgery, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226300, China;
| | - Jinzhu Wu
- Medical School, Nantong University, Nantong 226300, China; (S.Y.); (J.L.); (B.C.)
- Department of Hepatobiliary Surgery, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226300, China;
| |
Collapse
|
14
|
Guarnieri JW, Lie T, Albrecht YES, Hewin P, Jurado KA, Widjaja GA, Zhu Y, McManus MJ, Kilbaugh TJ, Keith K, Potluri P, Taylor D, Angelin A, Murdock DG, Wallace DC. Mitochondrial antioxidants abate SARS-COV-2 pathology in mice. Proc Natl Acad Sci U S A 2024; 121:e2321972121. [PMID: 39008677 PMCID: PMC11287122 DOI: 10.1073/pnas.2321972121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 06/07/2024] [Indexed: 07/17/2024] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection inhibits mitochondrial oxidative phosphorylation (OXPHOS) and elevates mitochondrial reactive oxygen species (ROS, mROS) which activates hypoxia-inducible factor-1alpha (HIF-1α), shifting metabolism toward glycolysis to drive viral biogenesis but also causing the release of mitochondrial DNA (mtDNA) and activation of innate immunity. To determine whether mitochondrially targeted antioxidants could mitigate these viral effects, we challenged mice expressing human angiotensin-converting enzyme 2 (ACE2) with SARS-CoV-2 and intervened using transgenic and pharmacological mitochondrially targeted catalytic antioxidants. Transgenic expression of mitochondrially targeted catalase (mCAT) or systemic treatment with EUK8 decreased weight loss, clinical severity, and circulating levels of mtDNA; as well as reduced lung levels of HIF-1α, viral proteins, and inflammatory cytokines. RNA-sequencing of infected lungs revealed that mCAT and Eukarion 8 (EUK8) up-regulated OXPHOS gene expression and down-regulated HIF-1α and its target genes as well as innate immune gene expression. These data demonstrate that SARS-CoV-2 pathology can be mitigated by catalytically reducing mROS, potentially providing a unique host-directed pharmacological therapy for COVID-19 which is not subject to viral mutational resistance.
Collapse
Affiliation(s)
- Joseph W. Guarnieri
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Timothy Lie
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- University of Pennsylvania, Philadelphia, PA19104
| | - Yentli E. Soto Albrecht
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Peter Hewin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Kellie A. Jurado
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Gabrielle A. Widjaja
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Yi Zhu
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Meagan J. McManus
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Todd J. Kilbaugh
- Department of Anesthesiology and Critical Care, Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Kelsey Keith
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Prasanth Potluri
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Deanne Taylor
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Department of Biomedical and Health Informatics, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Alessia Angelin
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
| | - Deborah G. Murdock
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Division of Human Genetics, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | - Douglas C. Wallace
- The Center for Mitochondrial and Epigenomic Medicine, The Children's Hospital of Philadelphia, Philadelphia, PA19104
- Division of Human Genetics, Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| |
Collapse
|
15
|
Papinska JA, Durślewicz J, Bagavant H, Deshmukh US. Deleting Mitochondrial Superoxide Dismutase 2 in Salivary Gland Ductal Epithelial Cells Recapitulates Non-Sjögren's Sicca Syndrome. Int J Mol Sci 2024; 25:5983. [PMID: 38892170 PMCID: PMC11172772 DOI: 10.3390/ijms25115983] [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: 04/09/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Elevated oxidative stress can play a pivotal role in autoimmune diseases by exacerbating inflammatory responses and tissue damage. In Sjögren's disease (SjD), the contribution of oxidative stress in the disease pathogenesis remains unclear. To address this question, we created mice with a tamoxifen-inducible conditional knockout (KO) of a critical antioxidant enzyme, superoxide dismutase 2 (Sod2), in the salivary glands (i-sg-Sod2 KO mice). Following tamoxifen treatment, Sod2 deletion occurred primarily in the ductal epithelium, and the salivary glands showed a significant downregulation of Sod2 expression. At twelve weeks post-treatment, salivary glands from the i-sg-Sod2 KO mice exhibited increased 3-Nitrotyrosine staining. Bulk RNA-seq revealed alterations in gene expression pathways related to ribosome biogenesis, mitochondrial function, and oxidative phosphorylation. Significant changes were noted in genes characteristic of salivary gland ionocytes. The i-sg-Sod2 KO mice developed reversible glandular hypofunction. However, this functional loss was not accompanied by glandular lymphocytic foci or circulating anti-nuclear antibodies. These data demonstrate that although localized oxidative stress in salivary gland ductal cells was insufficient for SjD development, it induced glandular dysfunction. The i-sg-Sod2 KO mouse resembles patients classified as non-Sjögren's sicca and will be a valuable model for deciphering oxidative-stress-mediated glandular dysfunction and recovery mechanisms.
Collapse
Affiliation(s)
- Joanna A. Papinska
- Department of Microbiology and Immunology, Oklahoma University Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Justyna Durślewicz
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.D.); (H.B.)
| | - Harini Bagavant
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.D.); (H.B.)
| | - Umesh S. Deshmukh
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK 73104, USA; (J.D.); (H.B.)
| |
Collapse
|
16
|
De I, Singh R, Kumar S, Singh S, Singh M, Panda JJ, Ghosh K, Mishra DP, Singh M. Short term biodistribution and in vivo toxicity assessment of intravenously injected pristine graphene oxide nanoflakes in SD rats. Toxicol Res (Camb) 2024; 13:tfae058. [PMID: 38617714 PMCID: PMC11014786 DOI: 10.1093/toxres/tfae058] [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: 08/22/2023] [Revised: 02/20/2024] [Accepted: 04/02/2024] [Indexed: 04/16/2024] Open
Abstract
The present study aimed to elucidate the short term biodistribution of nano sized graphene oxide (GO) along with the toxicological assessment under in-vivo condition with an intent to analyse the toxic effects of sudden accidental exposure of GO The synthesised GO was characterized using UV-Visible spectroscopy, XRD, FTIR, Raman spectroscopy, TGA and DLS. The morphological imaging was performed using SEM, TEM and AFM. With a lateral size of less than 300 nm, these nanoparticles exhibit significant organ barrier permeability of up to 20%. Upon acute exposure to 10 mg/kg dose of ICG-tagged GO nanoflakes through intravenous route, various organs such as kidney, spleen and liver were observed, and the nanoparticles predominantly accumulated in the liver upon 24 h of exposure. Upon confirming the accumulation of these particles in liver through IVIS imaging, our next attempt was to analyse various biochemical and serum parameters. An elevation in various serum parameters such as ALT, AST, Creatinine and Bilirubin was observed. Similarly, in the case of biochemical parameters tested in liver homogenates, an increase in NO, Catalase, GSH, SOD, ROS, LPO, GR, GPx, and GST was observed. This study highlights the potential toxicological risk associated with GO exposure which must be taken into account for any risk analysis associated with GO based consumer products and the occupational hazards.
Collapse
Affiliation(s)
- Indranil De
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, 140306, Mohali, Punjab, India
| | - Rashmika Singh
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, 140306, Mohali, Punjab, India
| | - Sushil Kumar
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, 140306, Mohali, Punjab, India
| | - Srishti Singh
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, 140306, Mohali, Punjab, India
| | - Manohar Singh
- CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Jiban Jyoti Panda
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, 140306, Mohali, Punjab, India
| | - Kaushik Ghosh
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, 140306, Mohali, Punjab, India
| | | | - Manish Singh
- Institute of Nano Science and Technology (INST), Knowledge City, Sector-81, SAS Nagar, 140306, Mohali, Punjab, India
| |
Collapse
|
17
|
Mogck BA, Jezak ST, Wiley CD. Mitochondria-Targeted Catalase Does Not Suppress Development of Cellular Senescence during Aging. Biomedicines 2024; 12:414. [PMID: 38398016 PMCID: PMC10886841 DOI: 10.3390/biomedicines12020414] [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: 12/19/2023] [Revised: 02/06/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
Cellular senescence is a complex stress response marked by stable proliferative arrest and the secretion of biologically active molecules collectively known as the senescence-associated secretory phenotype (SASP). Mitochondria-derived reactive oxygen species (ROS) have been implicated in aging and age-related processes, including senescence. Stressors that increase ROS levels promote both senescence and the SASP, while reducing mitochondrial ROS or mitochondria themselves can prevent senescence or the SASP. Mitochondrially targeted catalase (mCAT), a transgene that reduces mitochondrial levels of ROS, has been shown to extend the lifespan of murine models and protect against the age-related loss of mitochondrial function. However, it remains unclear whether mCAT can prevent senescence or the SASP. In this study, we investigated the impact of mCAT on senescence in cultured cells and aged mice in order to discover if the lifespan-extending activity of mCAT might be due to the reduction in senescent cells or the SASP. Contrary to expectations, we observed that mCAT does not reduce markers of senescence or the SASP in cultured cells. Moreover, mCAT does not prevent the accumulation of senescent cells or the development of the SASP in adipose tissue from aged mice. These results suggest that mitochondrial ROS may not always play a causal role in the development of senescence during natural aging and underscore the need for a nuanced understanding of the intricate relationship between mitochondrial ROS and cellular senescence.
Collapse
Affiliation(s)
- Bronwyn A. Mogck
- Jean Mayer USDA Human Nutrition Research on Aging, Tufts University, Boston, MA 02111, USA
- SENS Research Foundation, Mountain View, CA 94041, USA
| | - Samantha T. Jezak
- Jean Mayer USDA Human Nutrition Research on Aging, Tufts University, Boston, MA 02111, USA
- Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA 02111, USA
| | - Christopher D. Wiley
- Jean Mayer USDA Human Nutrition Research on Aging, Tufts University, Boston, MA 02111, USA
- Friedman School of Nutrition Science & Policy, Tufts University, Boston, MA 02111, USA
- Department of Medicine, School of Medicine, Tufts University, Boston, MA 02111, USA
| |
Collapse
|
18
|
Kishi S, Nagasu H, Kidokoro K, Kashihara N. Oxidative stress and the role of redox signalling in chronic kidney disease. Nat Rev Nephrol 2024; 20:101-119. [PMID: 37857763 DOI: 10.1038/s41581-023-00775-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/21/2023]
Abstract
Chronic kidney disease (CKD) is a major public health concern, underscoring a need to identify pathogenic mechanisms and potential therapeutic targets. Reactive oxygen species (ROS) are derivatives of oxygen molecules that are generated during aerobic metabolism and are involved in a variety of cellular functions that are governed by redox conditions. Low levels of ROS are required for diverse processes, including intracellular signal transduction, metabolism, immune and hypoxic responses, and transcriptional regulation. However, excess ROS can be pathological, and contribute to the development and progression of chronic diseases. Despite evidence linking elevated levels of ROS to CKD development and progression, the use of low-molecular-weight antioxidants to remove ROS has not been successful in preventing or slowing disease progression. More recent advances have enabled evaluation of the molecular interactions between specific ROS and their targets in redox signalling pathways. Such studies may pave the way for the development of sophisticated treatments that allow the selective control of specific ROS-mediated signalling pathways.
Collapse
Affiliation(s)
- Seiji Kishi
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Hajime Nagasu
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Kengo Kidokoro
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan
| | - Naoki Kashihara
- Department of Nephrology and Hypertension, Kawasaki Medical School, Kurashiki, Okayama, Japan.
| |
Collapse
|
19
|
Jin Y, Zhang J, Guo Q, Dong X, Li J, Wang J, Li S, Shen Y, Lin K, Yang Z, Chu J, Sun H, Luo Z. Exploring Genetic Diversity of SOD2 and POU5F1 for Congenital Heart Disease in the Southwest Chinese Population. Int Heart J 2024; 65:723-729. [PMID: 39085111 DOI: 10.1536/ihj.24-068] [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] [Indexed: 08/02/2024]
Abstract
Congenital heart disease (CHD) accounts for nearly one-third of all major congenital anomalies, with atrial septal defect (ASD) and ventricular septal defect (VSD) being the most common forms of simple CHD, which involve a large number of susceptibility genes. However, despite extensive research, the etiology of ASD and VSD remains unclear. Yunnan Province has advantages in exploring CHD pathogenesis due to its unique genetic background. Therefore, we aimed to evaluate the association between single nucleotide polymorphisms (SNPs) of genes and susceptibility to simple CHD in a specific population by means of a case-control study. A total of 337 healthy controls and 767 patients with simple CHD (501 ASD and 266 VSD) from China were recruited. Candidate SNPs were identified through whole-genome sequencing of pooled CHD patients and controls (pool-seq). Genotyping from 1,104 samples was performed, and stratified analysis was conducted to explore the association between positive SNPs and CHD subtypes. χ2 tests and logistic regression were used to analyze the relationship between each SNP and simple CHD. Of 11 SNPs identified, SOD2 rs62437333 (P = 0.005) and POU5F1 rs3130504 (P = 0.017) showed differences between the control and ASD cohorts. In the dominant inheritance model hypothesis, rs62437333 allele C carriers had increased ASD (odds ratio (OR) = 2.04, P = 0.005) and combined simple CHD risk (OR = 2.33, P = 0.012) compared to DD genotype, while rs3130504 allele C carriers had increased ASD risk (OR = 1.121, P = 0.045) compared to DD genotype.
Collapse
Affiliation(s)
- Ye Jin
- Yunnan Fuwai Cardiovascular Hospital
| | - Jun Zhang
- Yunnan Fuwai Cardiovascular Hospital
| | | | | | - Jian Li
- Yunnan Fuwai Cardiovascular Hospital
| | | | - Shuang Li
- Yunnan Fuwai Cardiovascular Hospital
| | - Yan Shen
- Yunnan Fuwai Cardiovascular Hospital
| | - Keqin Lin
- Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Zhaoqing Yang
- Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Jiayou Chu
- Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Hao Sun
- Department of Medical Genetics, Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College
| | | |
Collapse
|
20
|
Bir A, Ghosh A, Müller WE, Ganguly A. Mitochondrial dysfunction and metabolic syndrome. METABOLIC SYNDROME 2024:157-172. [DOI: 10.1016/b978-0-323-85732-1.00043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
21
|
Cui Y, Zhai Y, Yuan Y, Wang X, Xu Q, Wu X, Xu L, Ren T, Wang Q, Sun H. Inhibition of PTEN-induced kinase 1 autophosphorylation may assist in preventing epileptogenesis induced by pentylenetetrazol. Neurochem Int 2024; 172:105644. [PMID: 38029887 DOI: 10.1016/j.neuint.2023.105644] [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: 10/13/2023] [Revised: 11/15/2023] [Accepted: 11/21/2023] [Indexed: 12/01/2023]
Abstract
PTEN-induced kinase 1 (PINK1) autophosphorylation-triggered mitophagy is the main mitophagic pathway in the nervous system. Moreover, multiple studies have confirmed that mitophagy is closely related to the occurrence and development of epilepsy. Therefore, we speculated that the PINK1 autophosphorylation may be involved in epileptogenesis by mediating mitophagic pathway. This study aimed to explore the contribution of activated PINK1 to epileptogenesis induced by pentylenetetrazol (PTZ) in Sprague‒Dawley rats. During PTZ-induced epileptogenesis, the levels of phosphorylated PINK1 were increased, accompanied by elevated mitophagy, mitochondria oxidative stress and neuronal damage. After microRNA intervention targeting translocase outer mitochondrial membrane 7 (TOM7) or overlapping with the m-AAA protease 1 homolog (OMA1), the levels of PINK1 phosphorylation, mitophagy, mitochondrial oxidative stress, neuronal injury were observed in the rats with induced epileptogenesis. Furthermore, inhibiting of the expression of TOM7, a positive regulator of PINK1 autophosphorylation, reversed the increase in PINK1 phosphorylation and alleviated mitophagy, neuronal injury, thereby preventing epileptogenesis. In contrast, reducing the levels of OMA1, a negative regulator of PINK1 autophosphorylation, led to increased phosphorylation of PINK1, accompanied by aggravated neuronal injury and ultimately, epileptogenesis. This study confirmed the contribution of activated PINK1 to PTZ-induced epileptogenesis and suggested that the inhibition of PINK1 autophosphorylation may assist in preventing epileptogenesis.
Collapse
Affiliation(s)
- Yaru Cui
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yujie Zhai
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Yi Yuan
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xiaoqian Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Qianqian Xu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Xiangdong Wu
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Linlin Xu
- Department of Neurology, The Second Hospital of Shandong University, Jinan, 250033, China
| | - Tianpu Ren
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China
| | - Qiaoyun Wang
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| | - Hongliu Sun
- School of Pharmaceutical Sciences, Binzhou Medical University, Yantai, 264003, China.
| |
Collapse
|
22
|
Tavleeva MM, Rasova EE, Rybak AV, Belykh ES, Fefilova EA, Pnachina EM, Velegzhaninov IO. Dose-Dependent Effect of Mitochondrial Superoxide Dismutase Gene Overexpression on Radioresistance of HEK293T Cells. Int J Mol Sci 2023; 24:17315. [PMID: 38139144 PMCID: PMC10744337 DOI: 10.3390/ijms242417315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Over the last two decades, a multitude of gain-of-function studies have been conducted on genes that encode antioxidative enzymes, including one of the key enzymes, manganese superoxide dismutase (SOD2). The results of such studies are often contradictory, as they strongly depend on many factors, such as the gene overexpression level. In this study, the effect of altering the ectopic expression level of major transcript variants of the SOD2 gene on the radioresistance of HEK293T cells was investigated using CRISPRa technology. A significant increase in cell viability in comparison with the transfection control was detected in cells with moderate SOD2 overexpression after irradiation at 2 Gy, but not at 3 or 5 Gy. A further increase in the level of SOD2 ectopic expression up to 22.5-fold resulted in increased cell viability detectable only after irradiation at 5 Gy. Furthermore, a 15-20-fold increase in SOD2 expression raised the clonogenic survival of cells after irradiation at 5 Gy. Simultaneous overexpression of genes encoding SOD2 and Catalase (CAT) enhanced clonogenic cell survival after irradiation more effectively than separate overexpression of both. In conjunction with the literature data on the suppression of the procarcinogenic effects of superoxide dismutase overexpression by ectopic expression of CAT, the data presented here suggest the potential efficacy of simultaneous overexpression of SOD2 and CAT to reduce oxidative stress occurring in various pathological processes. Moreover, these results illustrate the importance of selecting the degree of SOD2 overexpression to obtain a protective effect.
Collapse
Affiliation(s)
- Marina M. Tavleeva
- Institute of Biology of Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia; (M.M.T.); (E.E.R.); (A.V.R.); (E.S.B.)
| | - Elena E. Rasova
- Institute of Biology of Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia; (M.M.T.); (E.E.R.); (A.V.R.); (E.S.B.)
| | - Anna V. Rybak
- Institute of Biology of Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia; (M.M.T.); (E.E.R.); (A.V.R.); (E.S.B.)
| | - Elena S. Belykh
- Institute of Biology of Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia; (M.M.T.); (E.E.R.); (A.V.R.); (E.S.B.)
| | - Elizaveta A. Fefilova
- Institute of Cytology, Russian Academy of Sciences, 4 Tikhoretsky Ave., St. Petersburg 194064, Russia;
| | - Elizaveta M. Pnachina
- Institute of Biology and Biomedicine, Lobachevsky State University of Nizhny Novgorod, 23 Gagarin Ave., Nizhny Novgorod 603950, Russia;
| | - Ilya O. Velegzhaninov
- Institute of Biology of Komi Scientific Centre, Ural Branch of Russian Academy of Sciences, 28b Kommunisticheskaya St., Syktyvkar 167982, Russia; (M.M.T.); (E.E.R.); (A.V.R.); (E.S.B.)
| |
Collapse
|
23
|
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
|
24
|
Haak JL, Kregel KC, Bloomer SA. Altered accumulation of hepatic mitochondrial antioxidant proteins with age and environmental heat stress. J Appl Physiol (1985) 2023; 135:1339-1347. [PMID: 37881850 PMCID: PMC10979832 DOI: 10.1152/japplphysiol.00610.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/20/2023] [Accepted: 10/20/2023] [Indexed: 10/27/2023] Open
Abstract
Aging impairs overall physiological function, particularly the response to environmental stressors. Repeated heat stress elevates reactive oxygen species and macromolecular damage in the livers of aged animals, likely due to mitochondrial dysfunction. The goal of this investigation was to determine potential mechanisms for mitochondrial dysfunction after heat stress by evaluating key redox-sensitive and antioxidant proteins (Sirt-3, MnSOD, Trx-2, and Ref-1). We hypothesized that heat stress would result in greater mitochondrial abundance of these proteins, but that aging would attenuate this response. For this purpose, young (6 mo) and old (24 mo) Fisher 344 rats were exposed to heat stress on two consecutive days. During each heating trial, colonic temperature was elevated to 41°C during the first 60 min, and then clamped at this temperature for 30 min. Nonheated animals served as controls. At 2 and 24 h after the second heat stress, hepatic mitochondria were isolated from each animal, and then immunoblotted for Sirt-3, acetylated lysine residues (Ac-K), MnSOD, Trx-2, and Ref-1. Aging increased Sirt-3 and lowered Ac-K. In response to heat stress, Sirt-3, Ac-K, MnSOD, and Ref-1 increased in mitochondrial fractions in both young and old animals. At 2 h after the second heat stress, mitochondrial Trx-2 declined in old, but not in young animals. Our results suggest that some components of the response to heat stress are preserved with aging. However, the decline in Trx-2 represents a potential mechanism for age-related mitochondrial damage and dysfunction after heat stress.NEW & NOTEWORTHY Our results suggest heat stress-induced mitochondrial translocation of Sirt-3, MnSOD, and Ref-1 in young and old animals. Aged rats experienced a decline in Trx-2 after heat stress, suggesting a potential mechanism for age-related mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jodie L Haak
- Health Sciences Department, Drexel University, Philadelphia, Pennsylvania, United States
| | - Kevin C Kregel
- Department of Health and Human Physiology, The University of Iowa, Iowa City, Iowa, United States
| | - Steven A Bloomer
- Division of Science and Engineering, Penn State Abington, Abington, Pennsylvania, United States
| |
Collapse
|
25
|
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
|
26
|
Kang R, Laborde C, Savchenko L, Swiader A, Pizzinat N, Marsal D, Sainte-Marie Y, Boal F, Tronchere H, Roncalli J, Kunduzova O. Age-Related Shift in Cardiac and Metabolic Phenotyping Linked to Inflammatory Cytokines and Antioxidant Status in Mice. Int J Mol Sci 2023; 24:15841. [PMID: 37958823 PMCID: PMC10650425 DOI: 10.3390/ijms242115841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/18/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Age-related alterations in cardiac function, metabolic, inflammatory and antioxidant profiles are associated with an increased risk of cardiovascular mortality and morbidity. Here, we examined cardiac and metabolic phenotypes in relation to inflammatory status and antioxidant capacity in young, middle-aged and old mice. Real-time reverse transcription-polymerase chain reactions were performed on myocardium and immunoassays on plasma. Left ventricular (LV) structure and function were assessed by echocardiography using high-frequency ultrasound. Middle-aged mice exhibited an altered metabolic profile and antioxidant capacity compared to young mice, whereas myocardial expression of inflammatory factors (TNFα, IL1β, IL6 and IL10) remained unchanged. In contrast, old mice exhibited increased expression of inflammatory cytokines and plasma levels of resistin compared to young and middle-aged mice (p < 0.05). The pro-inflammatory signature of aged hearts was associated with alterations in glutathione redox homeostasis and elevated contents of 4-hydroxynonenal (4-HNE), a marker of lipid peroxidation and oxidative stress. Furthermore, echocardiographic parameters of LV systolic and diastolic functions were significantly altered in old mice compared to young mice. Taken together, these findings suggest age-related shifts in cardiac phenotype encompass the spectrum of metabo-inflammatory abnormalities and altered redox homeostasis.
Collapse
Affiliation(s)
- Ryeonshi Kang
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | | | - Lesia Savchenko
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
- Department of Internal Medicine, Poltava State Medical University, 23 Shevchenko, 36000 Poltava, Ukraine
| | - Audrey Swiader
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Nathalie Pizzinat
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Dimitri Marsal
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Yannis Sainte-Marie
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Frederic Boal
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Helene Tronchere
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| | - Jerome Roncalli
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- Department of Cardiology, University Hospital of Toulouse, CEDEX 9, 31400 Toulouse, France
| | - Oksana Kunduzova
- National Institute of Health and Medical Research (INSERM) U1297, CEDEX 4, 31432 Toulouse, France; (R.K.); (L.S.); (A.S.); (N.P.); (D.M.); (Y.S.-M.); (F.B.); (H.T.); (J.R.)
- University of Toulouse III, CEDEX 9, 31062 Toulouse, France;
| |
Collapse
|
27
|
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
|
28
|
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
|
29
|
Liu W, Zhang Y, Wei G, Zhang M, Li T, Liu Q, Zhou Z, Du Y, Wei H. Integrated Cascade Nanozymes with Antisenescence Activities for Atherosclerosis Therapy. Angew Chem Int Ed Engl 2023; 62:e202304465. [PMID: 37338457 DOI: 10.1002/anie.202304465] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/19/2023] [Accepted: 06/20/2023] [Indexed: 06/21/2023]
Abstract
Senescent cells are the critical drivers of atherosclerosis formation and maturation. Mitigating senescent cells holds promise for the treatment of atherosclerosis. In an atherosclerotic plaque microenvironment, senescent cells interact with reactive oxygen species (ROS), promoting the disease development. Here, we hypothesize that a cascade nanozyme with antisenescence and antioxidant activities can serve as an effective therapeutic for atherosclerosis. An integrated cascade nanozyme with superoxide dismutase- and glutathione peroxidase-like activities, named MSe1 , is developed in this work. The obtained cascade nanozyme can attenuate human umbilical vein endothelial cell (HUVEC) senescence by protecting DNA from damage. It significantly weakens inflammation in macrophages and HUVECs by eliminating overproduced intracellular ROS. Additionally, the MSe1 nanozyme effectively inhibits foam cell formation in macrophages and HUVECs by decreasing the internalization of oxidized low-density lipoprotein. After intravenous administration, the MSe1 nanozyme significantly inhibits the formation of atherosclerosis in apolipoprotein E-deficient (ApoE-/- ) mice by reducing oxidative stress and inflammation and then decreases the infiltration of inflammatory cells and senescent cells in atherosclerotic plaques. This study not only provides a cascade nanozyme but also suggests that the combination of antisenescence and antioxidative stress holds considerable promise for treating atherosclerosis.
Collapse
Affiliation(s)
- Wanling Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210023, Nanjing, Jiangsu, China
| | - Yihong Zhang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210023, Nanjing, Jiangsu, China
| | - Gen Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210023, Nanjing, Jiangsu, China
| | - Minxuan Zhang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210023, Nanjing, Jiangsu, China
| | - Tong Li
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210023, Nanjing, Jiangsu, China
| | - Quanyi Liu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Jilin, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Zijun Zhou
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210023, Nanjing, Jiangsu, China
| | - Yan Du
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, 130022, Jilin, Changchun, China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, 230026, Hefei, Anhui, China
| | - Hui Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, 210023, Nanjing, Jiangsu, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, 210023, Nanjing, Jiangsu, China
| |
Collapse
|
30
|
Zhang P, Lu H, Wu Y, Lu D, Li C, Yang X, Chen Z, Qian J, Ge J. COX5A Alleviates Doxorubicin-Induced Cardiotoxicity by Suppressing Oxidative Stress, Mitochondrial Dysfunction and Cardiomyocyte Apoptosis. Int J Mol Sci 2023; 24:10400. [PMID: 37373547 DOI: 10.3390/ijms241210400] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/15/2023] [Accepted: 06/18/2023] [Indexed: 06/29/2023] Open
Abstract
Doxorubicin (DOX) as a chemotherapeutic agent can cause mitochondrial dysfunction and heart failure. COX5A has been described as an important regulator of mitochondrial energy metabolism. We investigate the roles of COX5A in DOX-induced cardiomyopathy and explore the underlying mechanisms. C57BL/6J mice and H9c2 cardiomyoblasts were treated with DOX, and the COX5A expression was assessed. An adeno-associated virus serum type 9 (AAV9) and lenti-virus system were used to upregulate COX5A expression. Echocardiographic parameters, morphological and histological analyses, transmission electron microscope and immunofluorescence assays were used to assess cardiac and mitochondrial function. In a human study, we found that cardiac COX5A expression was dramatically decreased in patients with end-stage dilated cardiomyopathy (DCM) compared to the control group. COX5A was significantly downregulated following DOX stimulation in the heart of mice and H9c2 cells. Reduced cardiac function, decreased myocardium glucose uptake, mitochondrial morphology disturbance, reduced activity of mitochondrial cytochrome c oxidase (COX) and lowered ATP content were detected after DOX stimulation in mice, which could be significantly improved by overexpression of COX5A. Overexpression of COX5A effectively protected against DOX-induced oxidative stress, mitochondrial dysfunction and cardiomyocyte apoptosis in vivo and in vitro. Mechanistically, the phosphorylation of Akt (Thr308) and Akt (Ser473) were also decreased following DOX treatment, which could be reserved by the upregulation of COX5A. Furthermore, PI3K inhibitors abrogated the protection effects of COX5A against DOX-induced cardiotoxicity in H9c2 cells. Thus, we identified that PI3K/Akt signaling was responsible for the COX5A-mediated protective role in DOX-induced cardiomyopathy. These results demonstrated the protective effect of COX5A in mitochondrial dysfunction, oxidative stress, and cardiomyocyte apoptosis, providing a potential therapeutic target in DOX-induced cardiomyopathy.
Collapse
Affiliation(s)
- Peipei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Hao Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Yuan Wu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Danbo Lu
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Chenguang Li
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Xiangdong Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Zhangwei Chen
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Juying Qian
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, National Clinical Research Center for Interventional Medicine, Shanghai 200032, China
| |
Collapse
|
31
|
Yang A, Guo L, Zhang Y, Qiao C, Wang Y, Li J, Wang M, Xing J, Li F, Ji L, Guo H, Zhang R. MFN2-mediated mitochondrial fusion facilitates acute hypobaric hypoxia-induced cardiac dysfunction by increasing glucose catabolism and ROS production. Biochim Biophys Acta Gen Subj 2023:130413. [PMID: 37331409 DOI: 10.1016/j.bbagen.2023.130413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/06/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND Rapid ascent to high-altitude environment which is characterized by acute hypobaric hypoxia (HH) may increase the risk of cardiac dysfunction. However, the potential regulatory mechanisms and prevention strategies for acute HH-induced cardiac dysfunction have not been fully clarified. Mitofusin 2 (MFN2) is highly expressed in the heart and is involved in the regulation of mitochondrial fusion and cell metabolism. To date, however, the significance of MFN2 in the heart under acute HH has not been investigated. METHODS AND RESULTS Our study revealed that MFN2 upregulation in hearts of mice during acute HH led to cardiac dysfunction. In vitro experiments showed that the decrease in oxygen concentration induced upregulation of MFN2, impairing cardiomyocyte contractility and increasing the risk of QT prolongation. Additionally, acute HH-induced MFN2 upregulation promoted glucose catabolism and led to excessive mitochondrial reactive oxygen species (ROS) production in cardiomyocytes, ultimately resulting in decreased mitochondrial function. Furthermore, co-immunoprecipitation (co-IP) and mass spectrometry analyses indicated that MFN2 interacted with the NADH-ubiquinone oxidoreductase 23 kDa subunit (NDUFS8). Specifically, acute HH-induced MFN2 upregulation increased NDUFS8-dependent complex I activity. CONCLUSIONS Taken together, our studies provide the first direct evidence that MFN2 upregulation exacerbates acute HH-induced cardiac dysfunction by increasing glucose catabolism and ROS production. GENERAL SIGNIFICANCE Our studies indicate that MFN2 may be a promising therapeutic target for cardiac dysfunction under acute HH.
Collapse
Affiliation(s)
- Ailin Yang
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Lifei Guo
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yanfang Zhang
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Chenjin Qiao
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Yijin Wang
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Jiaying Li
- College of Life Sciences, Northwest University, Xi'an 710069, China
| | - Min Wang
- College of Life Sciences, Northwest University, Xi'an 710069, China; Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jinliang Xing
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, China
| | - Fei Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Lele Ji
- Experimental Teaching Center of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Haitao Guo
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, China.
| | - Ru Zhang
- State Key Laboratory of Cancer Biology and Department of Physiology and Pathophysiology, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Tusa I, Menconi A, Tubita A, Rovida E. Pathophysiological Impact of the MEK5/ERK5 Pathway in Oxidative Stress. Cells 2023; 12:cells12081154. [PMID: 37190064 DOI: 10.3390/cells12081154] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/22/2023] [Accepted: 04/06/2023] [Indexed: 05/17/2023] Open
Abstract
Oxidative stress regulates many physiological and pathological processes. Indeed, a low increase in the basal level of reactive oxygen species (ROS) is essential for various cellular functions, including signal transduction, gene expression, cell survival or death, as well as antioxidant capacity. However, if the amount of generated ROS overcomes the antioxidant capacity, excessive ROS results in cellular dysfunctions as a consequence of damage to cellular components, including DNA, lipids and proteins, and may eventually lead to cell death or carcinogenesis. Both in vitro and in vivo investigations have shown that activation of the mitogen-activated protein kinase kinase 5/extracellular signal-regulated kinase 5 (MEK5/ERK5) pathway is frequently involved in oxidative stress-elicited effects. In particular, accumulating evidence identified a prominent role of this pathway in the anti-oxidative response. In this respect, activation of krüppel-like factor 2/4 and nuclear factor erythroid 2-related factor 2 emerged among the most frequent events in ERK5-mediated response to oxidative stress. This review summarizes what is known about the role of the MEK5/ERK5 pathway in the response to oxidative stress in pathophysiological contexts within the cardiovascular, respiratory, lymphohematopoietic, urinary and central nervous systems. The possible beneficial or detrimental effects exerted by the MEK5/ERK5 pathway in the above systems are also discussed.
Collapse
Affiliation(s)
- Ignazia Tusa
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessio Menconi
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Alessandro Tubita
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| | - Elisabetta Rovida
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, 50134 Florence, Italy
| |
Collapse
|
35
|
Chemical characterization of polysaccharides from Arctium lappa root and its hepatoprotective effects on mice. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
|
36
|
Castejon-Vega B, Cordero MD, Sanz A. How the Disruption of Mitochondrial Redox Signalling Contributes to Ageing. Antioxidants (Basel) 2023; 12:831. [PMID: 37107206 PMCID: PMC10135186 DOI: 10.3390/antiox12040831] [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: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023] Open
Abstract
In the past, mitochondrial reactive oxygen species (mtROS) were considered a byproduct of cellular metabolism. Due to the capacity of mtROS to cause oxidative damage, they were proposed as the main drivers of ageing and age-related diseases. Today, we know that mtROS are cellular messengers instrumental in maintaining cellular homeostasis. As cellular messengers, they are produced in specific places at specific times, and the intensity and duration of the ROS signal determine the downstream effects of mitochondrial redox signalling. We do not know yet all the processes for which mtROS are important, but we have learnt that they are essential in decisions that affect cellular differentiation, proliferation and survival. On top of causing damage due to their capacity to oxidize cellular components, mtROS contribute to the onset of degenerative diseases when redox signalling becomes dysregulated. Here, we review the best-characterized signalling pathways in which mtROS participate and those pathological processes in which they are involved. We focus on how mtROS signalling is altered during ageing and discuss whether the accumulation of damaged mitochondria without signalling capacity is a cause or a consequence of ageing.
Collapse
Affiliation(s)
- Beatriz Castejon-Vega
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Mario D. Cordero
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
- Department of Molecular Biology and Biochemical Engineering, Universidad Pablo de Olavide, 41013 Seville, Spain
| | - Alberto Sanz
- School of Molecular Biosciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| |
Collapse
|
37
|
Oh SH, Lee SE, Han DH, Yoon JW, Kim SH, Lim ES, Lee HB, Kim EY, Park SP. Treatments of Porcine Nuclear Recipient Oocytes and Somatic Cell Nuclear Transfer-Generated Embryos with Various Reactive Oxygen Species Scavengers Lead to Improvements of Their Quality Parameters and Developmental Competences by Mitigating Oxidative Stress-Related Impacts. Cell Reprogram 2023; 25:73-81. [PMID: 36939858 DOI: 10.1089/cell.2022.0145] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023] Open
Abstract
This study investigated the antioxidant effects of β-cryptoxanthin (BCX), hesperetin (HES), and icariin (ICA), and their effects on in vitro maturation of porcine oocytes and subsequent embryonic development of somatic cell nuclear transfer (SCNT). Treatment with 1 μM BCX (BCX-1) increased the developmental rate of porcine oocytes more than treatment with 100 μM HES (HES-100) or 5 μM ICA (ICA-5). The glutathione level and mRNA expression of antioxidant genes (NFE2L2, SOD1, and SOD2) were more increased in the BCX-1 group than in the HES-100 and ICA-5 groups, while the reactive oxygen species level was more decreased. Moreover, BCX improved the developmental capacity and quality of SCNT embryos. The total cell number, apoptotic cell rate, and development-related gene expression were modulated in the BCX-1 group to enhance embryonic development of SCNT. These results show that the antioxidant effects of BCX enhance in vitro maturation of porcine oocytes and subsequent embryonic development of SCNT.
Collapse
Affiliation(s)
- Seung-Hwan Oh
- Stem Cell Research Center, Jeju National University, Jeju, Korea
| | - Seung-Eun Lee
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Korea
| | - Dong-Hun Han
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Korea
| | - Jae-Wook Yoon
- Stem Cell Research Center, Jeju National University, Jeju, Korea
| | - So-Hee Kim
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Korea
| | - Eun-Seo Lim
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Korea
| | - Han-Bi Lee
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Korea
| | - Eun-Young Kim
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju, Korea.,Mirae Cell Bio, Seoul, Korea
| | - Se-Pill Park
- Stem Cell Research Center, Jeju National University, Jeju, Korea.,Mirae Cell Bio, Seoul, Korea.,Department of Bio Medical Informatics, College of Applied Life Sciences, Jeju National University, Jeju, Korea
| |
Collapse
|
38
|
Ahmed A, Syed JN, Chi L, Wang Y, Perez-Romero C, Lee D, Kocaqi E, Caballero A, Yang J, Escalante-Covarrubias Q, Ishimura A, Suzuki T, Aguilar-Arnal L, Gonzales GB, Kim KH, Delgado-Olguín P. KDM8 epigenetically controls cardiac metabolism to prevent initiation of dilated cardiomyopathy. NATURE CARDIOVASCULAR RESEARCH 2023; 2:174-191. [PMID: 38665902 PMCID: PMC11041705 DOI: 10.1038/s44161-023-00214-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 01/05/2023] [Indexed: 04/28/2024]
Abstract
Cardiac metabolism is deranged in heart failure, but underlying mechanisms remain unclear. Here, we show that lysine demethylase 8 (Kdm8) maintains an active mitochondrial gene network by repressing Tbx15, thus preventing dilated cardiomyopathy leading to lethal heart failure. Deletion of Kdm8 in mouse cardiomyocytes increased H3K36me2 with activation of Tbx15 and repression of target genes in the NAD+ pathway before dilated cardiomyopathy initiated. NAD+ supplementation prevented dilated cardiomyopathy in Kdm8 mutant mice, and TBX15 overexpression blunted NAD+-activated cardiomyocyte respiration. Furthermore, KDM8 was downregulated in human hearts affected by dilated cardiomyopathy, and higher TBX15 expression defines a subgroup of affected hearts with the strongest downregulation of genes encoding mitochondrial proteins. Thus, KDM8 represses TBX15 to maintain cardiac metabolism. Our results suggest that epigenetic dysregulation of metabolic gene networks initiates myocardium deterioration toward heart failure and could underlie heterogeneity of dilated cardiomyopathy.
Collapse
Affiliation(s)
- Abdalla Ahmed
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Jibran Nehal Syed
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Lijun Chi
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Yaxu Wang
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Carmina Perez-Romero
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Dorothy Lee
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Physiology, University of Toronto, Toronto, Ontario Canada
| | - Etri Kocaqi
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Amalia Caballero
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
| | - Jielin Yang
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
| | - Quetzalcoatl Escalante-Covarrubias
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Akihiko Ishimura
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Takeshi Suzuki
- Division of Functional Genomics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Lorena Aguilar-Arnal
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México City, México
| | - Gerard Bryan Gonzales
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Kyoung-Han Kim
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa and University of Ottawa Heart Institute, Ottawa, Ontario Canada
| | - Paul Delgado-Olguín
- Department of Translational Medicine, The Hospital for Sick Children, Toronto, Ontario Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario Canada
- Heart & Stroke Richard Lewar Centre of Excellence, Toronto, Ontario Canada
| |
Collapse
|
39
|
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
|
40
|
Domán A, Dóka É, Garai D, Bogdándi V, Balla G, Balla J, Nagy P. Interactions of reactive sulfur species with metalloproteins. Redox Biol 2023; 60:102617. [PMID: 36738685 PMCID: PMC9926313 DOI: 10.1016/j.redox.2023.102617] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Reactive sulfur species (RSS) entail a diverse family of sulfur derivatives that have emerged as important effector molecules in H2S-mediated biological events. RSS (including H2S) can exert their biological roles via widespread interactions with metalloproteins. Metalloproteins are essential components along the metabolic route of oxygen in the body, from the transport and storage of O2, through cellular respiration, to the maintenance of redox homeostasis by elimination of reactive oxygen species (ROS). Moreover, heme peroxidases contribute to immune defense by killing pathogens using oxygen-derived H2O2 as a precursor for stronger oxidants. Coordination and redox reactions with metal centers are primary means of RSS to alter fundamental cellular functions. In addition to RSS-mediated metalloprotein functions, the reduction of high-valent metal centers by RSS results in radical formation and opens the way for subsequent per- and polysulfide formation, which may have implications in cellular protection against oxidative stress and in redox signaling. Furthermore, recent findings pointed out the potential role of RSS as substrates for mitochondrial energy production and their cytoprotective capacity, with the involvement of metalloproteins. The current review summarizes the interactions of RSS with protein metal centers and their biological implications with special emphasis on mechanistic aspects, sulfide-mediated signaling, and pathophysiological consequences. A deeper understanding of the biological actions of reactive sulfur species on a molecular level is primordial in H2S-related drug development and the advancement of redox medicine.
Collapse
Affiliation(s)
- Andrea Domán
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary
| | - Éva Dóka
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary
| | - Dorottya Garai
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary,Kálmán Laki Doctoral School, University of Debrecen, 4012, Debrecen, Hungary
| | - Virág Bogdándi
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary
| | - György Balla
- Kálmán Laki Doctoral School, University of Debrecen, 4012, Debrecen, Hungary,Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary,ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, 4012, Debrecen, Hungary
| | - József Balla
- Kálmán Laki Doctoral School, University of Debrecen, 4012, Debrecen, Hungary,ELKH-UD Vascular Pathophysiology Research Group, 11003, University of Debrecen, 4012, Debrecen, Hungary,Department of Nephrology, Institute of Internal Medicine, Faculty of Medicine, University of Debrecen, 4012, Debrecen, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, 1122, Budapest, Hungary; Department of Anatomy and Histology, ELKH Laboratory of Redox Biology, University of Veterinary Medicine, 1078, Budapest, Hungary; Chemistry Institute, University of Debrecen, 4012, Debrecen, Hungary.
| |
Collapse
|
41
|
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
|
42
|
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
|
43
|
Theobroma cacao fortified-feed ameliorates potassium bromate-induced oxidative damage in male Wistar rat. Toxicol Rep 2023; 10:269-280. [PMID: 36876028 PMCID: PMC9976575 DOI: 10.1016/j.toxrep.2023.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/31/2023] [Accepted: 02/16/2023] [Indexed: 02/19/2023] Open
Abstract
Some therapeutic and beneficial health properties of the Theobroma cacao leaf have been documented. This study evaluated the ameliorative effect of Theobroma cacao-fortified feed against potassium bromate-induced oxidative damage in male Wistar rats. Thirty rats were randomly grouped into A-E. Except for E (the negative control), the rats in the other groups were administered 0.5 ml of 10 mg/kg body weight of potassium bromate daily using oral gavage and then allowed access to feed and water ad libitum. Groups B, C, and D were fed with 10 %, 20 %, and 30 % leaf-fortified feed respectively, while the negative and positive control (A) was fed with commercial feed. The treatment was carried out consecutively for fourteen days. In the liver and kidney, there was a significant increase (p < 0.05) in total protein concentration, a significant decrease (P < 0.05) in MDA level, and SOD activity in the fortified feed group compared to the positive control. Furthermore, in the serum, there was a significant increase (p < 0.05) in the albumin concentration, and ALT activity, and a significant decrease (p < 0.05) in urea concentration in the fortified feed groups compared to the positive control. The histopathology of the liver and kidney in the treated groups showed moderate cell degeneration compared to the positive control group. Antioxidant activity due to the presence of flavonoids and metal chelating activity of fiber in Theobroma cacao leaf could be responsible for the ameliorative effect of the fortified feed against potassium bromate-induced oxidative damage.
Collapse
|
44
|
Sonkar VK, Eustes AS, Ahmed A, Jensen M, Solanki MV, Swamy J, Kumar R, Fidler TP, Houtman JC, Allen BG, Spitz DR, Abel ED, Dayal S. Endogenous SOD2 (Superoxide Dismutase) Regulates Platelet-Dependent Thrombin Generation and Thrombosis During Aging. Arterioscler Thromb Vasc Biol 2023; 43:79-91. [PMID: 36325902 PMCID: PMC9780178 DOI: 10.1161/atvbaha.121.317735] [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: 06/29/2022] [Accepted: 10/24/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND Reactive oxygen species (ROS) contribute to platelet hyperactivation during aging. Several oxidative pathways and antioxidant enzymes have been implicated; however, their mechanistic contributions during aging remain elusive. We hypothesized that mitochondria are an important source of platelet ROS and that mitochondrial SOD2 (superoxide dismutase) protects against mitochondrial ROS-driven platelet activation and thrombosis during aging. METHODS We studied littermates of platelet-specific SOD2-knockout (SOD2fl/flPf4Cre, pSOD2-KO) and control (SOD2fl/fl) mice at young (4-5 months) or old (18-20 months) ages. We examined agonist-induced platelet activation, platelet-dependent thrombin generation potential, and susceptibility to in vivo thrombosis. RESULTS Platelet αIIbβ3 activation, aggregation, and adhesion were increased to similar extents in aged mice of both genotypes compared with young mice. In contrast, the age-dependent increases in mitochondrial and total cellular ROS, calcium elevation, and phosphatidylserine exposure were augmented in platelets from pSOD2-KO mice compared with control mice. Aged pSOD2-KO mice showed increased platelet-dependent thrombin generation compared with aged control mice. In vivo, aged pSOD2-KO mice exhibited enhanced susceptibility to carotid artery and pulmonary thrombosis compared to aged control mice. Adoptive transfer of platelets from aged pSOD2-KO but not aged control mice increased thrombotic susceptibility in aged host mice, suggesting a prothrombotic effect of platelet pSOD2 deficiency. Treatment with avasopasem manganese (GC4419), a SOD mimetic, decreased platelet mitochondrial pro-oxidants, cellular ROS levels, and inhibited procoagulant platelet formation and arterial thrombosis in aged mice. CONCLUSIONS Platelet mitochondrial ROS contributes to age-related thrombosis and endogenous SOD2 protects from platelet-dependent thrombin generation and thrombosis during aging.
Collapse
Affiliation(s)
- Vijay K Sonkar
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Alicia S Eustes
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Azaj Ahmed
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Melissa Jensen
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Mitali V Solanki
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jagadish Swamy
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Rahul Kumar
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Trevor P. Fidler
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Jon C.D. Houtman
- Departments of Microbiology & Immunology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Bryan G. Allen
- Free Radical and Radiation Biology Program, Radiation Oncology, Holden Comprehensive Cancer Center University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Radiation Oncology, Holden Comprehensive Cancer Center University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - E. Dale Abel
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Current address David Geffen School of Medicine, UCLA, Los Angeles, CA
| | - Sanjana Dayal
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa City VA Healthcare System, Iowa City, Iowa
| |
Collapse
|
45
|
Hussain T, Kandeel M, Metwally E, Murtaza G, Kalhoro DH, Yin Y, Tan B, Chughtai MI, Yaseen A, Afzal A, Kalhoro MS. Unraveling the harmful effect of oxidative stress on male fertility: A mechanistic insight. Front Endocrinol (Lausanne) 2023; 14:1070692. [PMID: 36860366 PMCID: PMC9968806 DOI: 10.3389/fendo.2023.1070692] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Accepted: 01/02/2023] [Indexed: 02/16/2023] Open
Abstract
Male infertility is a widely debated issue that affects males globally. There are several mechanisms involved. Oxidative stress is accepted to be the main contributing factor, with sperm quality and quantity affected by the overproduction of free radicals. Excess reactive oxygen species (ROS) cannot be controlled by the antioxidant system and, thus, potentially impact male fertility and hamper sperm quality parameters. Mitochondria are the driving force of sperm motility; irregularities in their function may lead to apoptosis, alterations to signaling pathway function, and, ultimately, compromised fertility. Moreover, it has been observed that the prevalence of inflammation may arrest sperm function and the production of cytokines triggered by the overproduction of ROS. Further, oxidative stress interacts with seminal plasma proteomes that influence male fertility. Enhanced ROS production disturbs the cellular constituents, particularly DNA, and sperms are unable to impregnate the ovum. Here, we review the latest information to better understand the relationship between oxidative stress and male infertility, the role of mitochondria, the cellular response, inflammation and fertility, and the interaction of seminal plasma proteomes with oxidative stress, as well as highlight the influence of oxidative stress on hormones; collectively, all of these factors are assumed to be important for the regulation of male infertility. This article may help improve our understanding of male infertility and the strategies to prevent it.
Collapse
Affiliation(s)
- Tarique Hussain
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
- Animal Sciences Division, Nuclear Institute for Agriculture and Biology College (NIAB-C), Pakistan Institute of Engineering and Applied Sciences (PIEAS), Faisalabad, Pakistan
- *Correspondence: Tarique Hussain, ; Bie Tan,
| | - Mahmoud Kandeel
- Department of Biomedical Sciences, College of Veterinary Medicine, King Faisal University, Al-Hofuf, Al-Ahsa, Saudi Arabia
- Department of Pharmacology, Faculty of Veterinary Medicine, Kafrelshikh University, Kafrelshikh, Egypt
| | - Elsayed Metwally
- Department of Cytology and Histology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia, Egypt
| | - Ghulam Murtaza
- Department of Animal Reproduction, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tandojam, Sindh, Pakistan
| | - Dildar Hussain Kalhoro
- Department of Veterinary Microbiology, Faculty of Animal Husbandry and Veterinary Sciences, Sindh Agriculture University, Tandojam, Sindh, Pakistan
| | - Yulong Yin
- Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Changsha, Hunan, China
- *Correspondence: Tarique Hussain, ; Bie Tan,
| | - Muhammad Ismail Chughtai
- Animal Sciences Division, Nuclear Institute for Agriculture and Biology College (NIAB-C), Pakistan Institute of Engineering and Applied Sciences (PIEAS), Faisalabad, Pakistan
| | - Anjaleena Yaseen
- Animal Sciences Division, Nuclear Institute for Agriculture and Biology College (NIAB-C), Pakistan Institute of Engineering and Applied Sciences (PIEAS), Faisalabad, Pakistan
| | - Ali Afzal
- Department of Zoology, Minhaj University, Lahore, Pakistan
| | - Muhammad Saleem Kalhoro
- Food Engineering and Bioprocess Technology, Asian Institute of Technology, Bangkok, Thailand
| |
Collapse
|
46
|
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
|
47
|
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
|
48
|
Kumari M, Bajad SM, Kshirsagar SR, Chinde S, Balaji AS, Jerald Mahesh Kumar M, Saxena S, Kumari SI. Sub-chronic oral toxicity evaluation of herbo-metallic formulation Arshakuthar rasa in rats. JOURNAL OF ETHNOPHARMACOLOGY 2022; 298:115306. [PMID: 35443217 DOI: 10.1016/j.jep.2022.115306] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/15/2022] [Accepted: 04/13/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Arshakuthar rasa (AR) is a mercury based Ayurvedic herbo-metallic formulation. The concerns are being raised about the probable toxicity of mercury after prolonged use of AR. Hence, there is need for a long-term repeated in vivo toxicity study. The study will provide data with scientific evidence to enable safe use of the drug. Moreover, lack of toxicity study with AR incited us to perform sub-chronic study on rats. AIM OF THE STUDY The aim of the study is to generate data by performing a sub-chronic study to assess the toxicity of AR after its prolonged oral intake. MATERIALS AND METHODS The female and male rats were administered with 30 (low), 300 (medium) and 600 mg/kg BW/day (high) dose of AR for 90 consecutive days. The body weight, feed consumption and water intake were monitored weekly. On 91st day, blood was collected from retro-orbital plexus of rats and then sacrificed to harvest the vital organs for biochemical, haematological, histopathological, genotoxicity along with the expression study of oxidative stress related genes and the biodistribution of elements in the blood. RESULTS Significant alterations in serum biochemical parameters were observed at the medium and high doses. The histopathological changes were in corroboration with biochemical changes at high dose in liver. There was no detectable level of mercury in blood, less to moderate biochemical changes, no haematological changes, moderate regulation of stress-related genes, and low genotoxicity. These results indicated that AR can be considered as moderately toxic above 600 mg/kg BW and mildly toxic at 300 mg/kg BW. CONCLUSIONS It may be interpreted that AR may not induce grave toxic response in human after long-duration of oral administration at therapeutic doses.
Collapse
Affiliation(s)
- Monika Kumari
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, Telangana, India
| | - Shatrughna Madhukar Bajad
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shripad Rajendra Kshirsagar
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Srinivas Chinde
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, Telangana, India
| | - Andugulapati Sai Balaji
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, Telangana, India
| | - M Jerald Mahesh Kumar
- Animal House Division, CSIR-Centre for Cellular and Molecular Biology, Tarnaka, Hyderabad, 500007, Telangana, India
| | - Saileshnath Saxena
- Department of Rasa Sastra & Bhaishajya Kalpana, Dr. B.R.K.R. Govt. Ayurvedic College, Erragadda, Hyderabad, 500 038, Telangana, India
| | - Srinivas Indu Kumari
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Hyderabad, 500 007, Telangana, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
49
|
Peng F, Jiang D, Xu W, Sun Y, Zha Z, Tan X, Yu J, Pan C, Zheng Q, Chen W. AMPK/MFF Activation: Role in Mitochondrial Fission and Mitophagy in Dry Eye. Invest Ophthalmol Vis Sci 2022; 63:18. [PMID: 36374514 PMCID: PMC9669805 DOI: 10.1167/iovs.63.12.18] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022] Open
Abstract
Purpose To assess the role of mitochondrial morphology and adenosine monophosphate-activated protein kinase (AMPK)/mitochondrial fission factor (MFF) in dry eye and the underlying mechanisms. Methods Immortalized human corneal epithelial cells (HCECs) and primary HCECs were cultured under high osmotic pressure (HOP). C57BL/6 female mice were injected subcutaneously with scopolamine. Quantitative real-time PCR was used to measure mRNA expression. Protein expression was assessed by western blot and immunofluorescence staining. Mitochondrial morphology was observed by confocal microscopy and transmission electron microscopy. Results First, HOP induced mitochondrial oxidative damage to HCECs, accompanied by mitochondrial fission and increased mitophagy. Then, AMPK/MFF pathway proteins were increased consequent to HOP-induced energy metabolism dysfunction. Interestingly, the AMPK pathway promoted mitochondrial fission and mitophagy by increasing the recruitment of dynamin-related protein 1 (DRP1) to the mitochondrial outer membrane in the HOP group. Moreover, AMPK knockdown attenuated mitochondrial fission and mitophagy due to HOP in HCECs. AMPK activation triggered mitochondrial fission and mitophagy. Mitochondrial fission of HCECs stressed by HOP was mediated via MFF phosphorylation. MFF knockdown reversed mitochondrial fragmentation and mitophagy in HCECs treated with HOP. Inhibition of MFF protected HCECs against oxidative damage, cell death, and inflammation in the presence of HOP. Finally, we detected mitochondrial fission and AMPK pathway activation in vivo. Conclusions The AMPK/MFF pathway mediates the development of dry eye by positively regulating mitochondrial fission and mitophagy. Inhibition of mitochondrial fission can alleviate oxidative damage and inflammation in dry eye and may provide experimental evidence for treating dry eye.
Collapse
Affiliation(s)
- Fangli Peng
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dan Jiang
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Xu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yining Sun
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiwei Zha
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiying Tan
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jinjie Yu
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chengjie Pan
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qinxiang Zheng
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wei Chen
- School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
50
|
Siokas V, Stamati P, Pateraki G, Liampas I, Aloizou AM, Tsirelis D, Nousia A, Sgantzos M, Nasios G, Bogdanos DP, Dardiotis E. Analysis of SOD2 rs4880 Genetic Variant in Patients with Alzheimer's Disease. Curr Issues Mol Biol 2022; 44:4406-4414. [PMID: 36286017 PMCID: PMC9600469 DOI: 10.3390/cimb44100302] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 09/16/2022] [Accepted: 09/16/2022] [Indexed: 11/24/2022] Open
Abstract
A few gene loci that contribute to Alzheimer's Disease (AD) onset have been identified. Few studies have been published about the relationship between SOD2 rs4880 single nucleotide variant and AD, revealing inconsistent results. Therefore, the aim of the current study is to further examine the role of the SOD2 rs4880 in AD. We performed a case-control study with a total of 641 subjects (320 patients with probable AD, and 321 healthy controls). The statistical analysis was performed assuming five genetic models. The threshold for statistical significance was set at 0.05. The results revealed no association between SOD2 rs4880 and AD in any of the assumed genetic models that were examined [log-additive OR = 0.95 (0.76-1.19), over-dominant OR = 1.15 (0.85-1.57), recessive OR = 0.85 (0.59-1.22), dominant OR = 1.03 (0.72-1.47), and co-dominant OR1 = 1.10 (0.75-1.60) and OR2 = 0.90 (0.58-1.40)]. Adjustment for sex and subgroup analyses based on sex did not reveal any statistically significant results either. Based on our findings, SOD2 rs4880 does not appear to play a determining role in the risk of developing AD. Larger studies are warranted to elucidate the connection between rs4880 and AD.
Collapse
Affiliation(s)
- Vasileios Siokas
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Polyxeni Stamati
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Georgia Pateraki
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Ioannis Liampas
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Athina-Maria Aloizou
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Daniil Tsirelis
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Anastasia Nousia
- Department of Speech and Language Therapy, University of Ioannina, 45500 Ioannina, Greece
| | - Markos Sgantzos
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Grigorios Nasios
- Department of Speech and Language Therapy, University of Ioannina, 45500 Ioannina, Greece
| | - Dimitrios P. Bogdanos
- Department of Rheumatology and Clinical Immunology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Efthimios Dardiotis
- Laboratory of Neurogenetics, Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| |
Collapse
|